Sélection de la langue

Search

Sommaire du brevet 2695514 

Énoncé de désistement de responsabilité concernant l'information provenant de tiers

Une partie des informations de ce site Web a été fournie par des sources externes. Le gouvernement du Canada n'assume aucune responsabilité concernant la précision, l'actualité ou la fiabilité des informations fournies par les sources externes. Les utilisateurs qui désirent employer cette information devraient consulter directement la source des informations. Le contenu fourni par les sources externes n'est pas assujetti aux exigences sur les langues officielles, la protection des renseignements personnels et l'accessibilité.

Disponibilité de l'Abrégé et des Revendications

L'apparition de différences dans le texte et l'image des Revendications et de l'Abrégé dépend du moment auquel le document est publié. Les textes des Revendications et de l'Abrégé sont affichés :

  • lorsque la demande peut être examinée par le public;
  • lorsque le brevet est émis (délivrance).
(12) Demande de brevet: (11) CA 2695514
(54) Titre français: REGIONS ULTRA-CONSERVEES CODANT POUR DES ARNNC
(54) Titre anglais: ULTRACONSERVED REGIONS ENCODING NCRNAS
Statut: Réputée abandonnée et au-delà du délai pour le rétablissement - en attente de la réponse à l’avis de communication rejetée
Données bibliographiques
(51) Classification internationale des brevets (CIB):
  • C12N 15/113 (2010.01)
  • A61K 31/7105 (2006.01)
  • A61K 48/00 (2006.01)
  • A61P 35/00 (2006.01)
  • C12Q 1/00 (2006.01)
  • C40B 30/04 (2006.01)
  • G1N 33/53 (2006.01)
  • G1N 33/574 (2006.01)
(72) Inventeurs :
  • CROCE, CARLO M. (Etats-Unis d'Amérique)
(73) Titulaires :
  • THE OHIO STATE UNIVERSITY RESEARCH FOUNDATION
(71) Demandeurs :
  • THE OHIO STATE UNIVERSITY RESEARCH FOUNDATION (Etats-Unis d'Amérique)
(74) Agent: SMART & BIGGAR LP
(74) Co-agent:
(45) Délivré:
(86) Date de dépôt PCT: 2008-08-04
(87) Mise à la disponibilité du public: 2009-02-12
Requête d'examen: 2013-06-10
Licence disponible: S.O.
Cédé au domaine public: S.O.
(25) Langue des documents déposés: Anglais

Traité de coopération en matière de brevets (PCT): Oui
(86) Numéro de la demande PCT: PCT/US2008/072081
(87) Numéro de publication internationale PCT: US2008072081
(85) Entrée nationale: 2010-02-03

(30) Données de priorité de la demande:
Numéro de la demande Pays / territoire Date
60/963,329 (Etats-Unis d'Amérique) 2007-08-03

Abrégés

Abrégé français

La présente invention a pour objet des procédés de différenciation des cancers humains comprenant l'utilisation d'un ou plusieurs profils d'expression de régions ultra-conservées transcrites (T-UCR). L'association entre la localisation génomique des UCR et les éléments génomiques liés au cancer analysé est hautement statistiquement significative et comparable à celle rapportée pour les miARN.


Abrégé anglais


Described herein are methods for differentiate human cancers comprising using
one or more transcribed
ultracon-served regions(T-UCR) expression profiles where the association
between the genomic location of UCRs and the analyzed
can-cer-related genomic elements is highly statistically significant and
comparable to that reported for miRNAs.

Revendications

Note : Les revendications sont présentées dans la langue officielle dans laquelle elles ont été soumises.


CLAIMS
What is claimed is:
1. A method for differentiate human cancers comprising using one or more
transcribed ultraconserved regions(T-UCR) expression profiles wherein the
association
between the genomic location of UCRs and the analyzed cancer-related genomic
elements is
highly statistically significant and comparable to that reported for miRNAs.
2. The method of claim 1, wherein one or more T-UCRs differentially expressed
in human cancers are located in cancer associated genomic regions (CAGR)
specifically
associated with that type of cancer.
3. The method of claim 1, wherein a cluster of seven UCRs (uc.347 to uc.353)
are located within the CAGR.
4. The method claim 3, wherein, two UCRs: uc.349A(P) and uc.352(N) are
among the T-UCRs that are differentially expressed between normal and
malignant B-CLL
CD5 positive cells.
5. The method of claim 4, wherein one or more of the UCRs are located in
genomic regions altered during the malignant process.
6. The method of claim 1, wherein one or more T-UCRs are candidate genes for
cancer susceptibility.
7. The method of claim 1, wherein a signature of five UCRs, uc.269A(N),
uc.160(N), uc.215(N), uc.346A(P) and uc.348(N), is used to differentiate
between two CLL
prognosis groups.
8. The method of claim 1, wherein three out of the 5 UCRs have significant
antisense complementarity with 5 out of the 13 miRNAs from the signature,
giving rise to six
92

possible interacting pairs: uc.160:: rniR-24, uc.160:: miR-155, uc.160:: miR-
223,
uc.160::miR-146a, uc.346A::miR-155, and uc.-348::miR-29b.
9. The method of claim 8, wherein the UCRs are located in genomic regions
targeted during the malignant process and are indicative of putative
involvement in human
tumorigenesis.
10. A cDNA corresponding to uc.246(E).
11. The cDNA of claim 10 cloned and expressed by standard methods.
12. A cDNA corresponding to uc.269A(N).
13. The cDNA of claim 12 cloned and expressed by standard methods.
14. A method of claim 1, wherein the T-UCR has an oncogenic function for
uc.73(P) in colon cancer, wherein diminution of its over-expression induces
apoptosis and
has antiproliferative effects in colon cancer cells abnormally expressing the
T-UCR.
15. A method for assessing a functional regulatory pathway in which two or
more
types of ncRNAs interact and influence the phenotype comprising determining,
correlations
between the expression of UCRs and miRNAs in a cancer patient.
16. The method of claim 15, wherein the cancer is CLL
17. The method of claim 16, wherein the correlation comprises the existence of
the miRNA::T-UCR interaction.
18. The method of claim 15, wherein one or more nc-UCGs are altered at the
genomic level in a cancer patient.
19. The method of claim 18, wherein the cancer patient has leukemia
93

20. A model in which both coding and non-coding genes are involved in and
cooperate in human tumorigenesis comprising one or more UCRs .
21. A method of diagnosing whether a subject has, or is at risk for
developing, a
cancer linked to a cancer-associated chromosomal feature, comprising
evaluating the status in
the subject of at least one UCR gene located in close proximity to the cancer-
associated
chromosomal feature, by:
(i) measuring in a test sample from said subject the level of at least one UCR
gene
product from the UCR gene in the test sample, wherein an alteration in the
level of UCR gene
product in the test sample relative to the level of corresponding UCR gene
product in a
control sample is indicative of the subject either having, or being at risk
for developing, the
cancer;
(ii) analyzing the at least one UCR gene in the test sample for a deletion,
mutation or
amplification, wherein detection of a deletion, mutation and/or amplification
in the UCR gene
as compared to the corresponding UCR gene in the control sample is indicative
of the subject
either having, or being at risk for developing, the cancer; and/or
(iii) measuring the copy number of the at least one UCR gene in the test
sample,
wherein a copy number other than two for an UCR gene on a somatic chromosome
or sex
chromosome in a female, or other than one for an UCR gene on a sex chromosome
in a male,
is indicative of the subject either having, or being at risk for developing,
the cancer.
22. The method of claim 21, wherein the cancer-associated chromosomal feature
is selected from the group consisting of: a cancer-associated genomic region;
a chromosomal
fragile site; a human papillomavirus integration site; and a homeobox gene or
gene cluster.
23. The method of claim 1, wherein the cancer is selected from the group
consisting of: bladder cancer; esophageal cancer; lung cancer; stomach cancer;
kidney cancer;
cervical cancer; ovarian cancer; breast cancer; lymphoma; Ewing sarcoma;
hematopoietic
tumors; solid tumors; gastric cancer; colorectal cancer; brain cancer;
epithelial cancer;
nasopharyngeal cancer; uterine cancer; hepatic cancer; head-and-neck cancer;
renal cancer;
male germ cell tumors; malignant mesothelioma; myelodysplastic syndrome;
pancreatic or
biliary cancer; prostate cancer; thyroid cancer; urothelial cancer; renal
cancer; Wilm's tumor;
small cell lung cancer; melanoma; skin cancer; osteosarcoma; neuroblastoma;
leukemia
94

(acute lymphocytic leukemia, acute myeloid leukemia, chronic lymphocytic
leukemia);
glioblastoma multiforme; medulloblastoma; lymphoplasmacytoid lymphoma; and
rhabdomyosarcoma.
24. The method of claim 1, wherein the cancer-associated chromosomal feature
is
a chromosomal fragile site.
25. The method of claim 10, wherein the UCR gene is selected from the group
consisting of: a cluster of seven UCRs (uc.347 to uc.353); and combinations
thereof.
26. The method of claim 17, wherein the cancer is a leukemia.
27. A method of diagnosing whether a subject has, or is at risk for
developing, a
cancer, comprising: (1) reverse transcribing RNA from a test sample obtained
from the
subject to provide a set of target oligodeoxynucleotides; (2) hybridizing the
target
oligodeoxynucleotides to a microarray comprising one or more UCR specific
probe
oligonucleotides to provide a hybridization profile for the test sample; and
(3) comparing the
test sample hybridization profile to a hybridization profile generated from a
control sample,
wherein an alteration in the signal is indicative of the subject either
having, or being at risk
for developing, the cancer.
28. The method of claim 27, wherein the cancer is a cancer associated with a
cancer-associated chromosomal feature.
29. The method of claim 27, wherein the cancer is B-cell chronic lymphocytic
leukemia.
30. A method of diagnosing whether a subject has, or is at risk for
developing, a
cancer associated with one or more adverse prognostic markers in a subject,
comprising: (1)
reverse transcribing RNA from a test sample obtained from the subject to
provide a set of
target oligodeoxynucleotides; (2) hybridizing the target oligodeoxynucleotides
to a
microarray comprising one or more UCR-specific probe oligonucleotides to
provide a
hybridization profile for said test sample; and (3) comparing the test sample
hybridization

profile to a hybridization profile generated from a control sample, wherein an
alteration in the
signal is indicative of the subject either having, or being at risk for
developing, the cancer.
31. The method of claim 30, wherein the cancer is B-cell chronic lymphocytic
leukemia.
32. A method of diagnosing whether a subject has, or is at risk for
developing, a
cancer, comprising analyzing in a test sample from said subject at least one
UCR gene or
gene product associated with a cancer, wherein detection of a mutation in the
UCR gene or
gene product, as compared to the corresponding UCR gene or gene product in a
control
sample, is indicative of the subject having, or being at risk for developing,
the cancer.
33. The method of claim 46, wherein the at least one miR gene or gene product
is
a miR gene selected from the group consisting wherein the UCR gene is selected
from the
group consisting of: a cluster of seven UCRs (uc.347 to uc.353); and
combinations thereof
34. The method of claim 33, wherein the cancer is B-cell chronic lymphocytic
leukemia.
35. A pharmaceutical composition comprising a UCR gene product and a
pharmaceutically-acceptable carrier, wherein the isolated UCR gene product is
from a UCR
gene located in close proximity to a cancer-associated chromosomal feature.
36. The pharmaceutical composition of claim 35, wherein the cancer-associated
chromosomal feature is selected from the group consisting of a cancer-
associated genomic
region, and a chromosomal fragile site.
37. A pharmaceutical composition comprising a nucleic acid encoding an
isolated
UCR gene product from a UC gene located in close proximity to a cancer-
associated
chromosomal feature, and a pharmaceutically-acceptable carrier.
38. The pharmaceutical composition of claim 37, wherein the cancer-associated
chromosomal feature is selected from the group consisting of a cancer-
associated genomic
96

region, a chromosomal fragile site, a human papillomavirus integration site,
and a homeobox
gene or gene cluster
39. A method of treating cancer in a subject, comprising:
(1) providing a subject who has a cancer associated with a cancer-associated
chromosomal feature, in which at least one isolated UCR gene product from a
UCR gene
located in close proximity to the cancer-associated chromosomal feature is
down-regulated or
up-regulated in cancer cells of the subject as compared to control cells; and
(2) (a) when the at least one isolated miR gene product is down-regulated in
the cancer cells, administering to the subject an effective amount of at least
one isolated miR
gene product from the at least one UCR gene, such that proliferation of cancer
cells in the
subject is inhibited; or (b) when the at least one isolated UCR gene product
is up-regulated in
the cancer cells, administering to the subject an effective amount of at least
one compound
for inhibiting expression of the at least one UCR gene, such that
proliferation of cancer cells
in the subject is inhibited.
40. The method of claim 39, wherein the cancer-associated chromosomal feature
is selected from the group consisting of a cancer-associated genomic region,
and a
chromosomal fragile site.
41. The method of treating cancer associated with a cancer-associated
chromosomal feature, comprising:
(1) determining the amount of UC gene product expressed from at least one UCR
gene located in close proximity to the cancer-associated chromosomal feature
in cancer cells
from a subject, relative to control cells; and
(2) altering the amount of UC gene product expressed in the cancer cells by:
(i) administering to the subject an effective amount of at least one isolated
miR gene product from the UCR gene, if the amount of the UCR gene product
expressed in
the cancer cells is less than amount of the miR gene product expressed in
control cells; or
(ii) administering to the subject an effective amount of at least one compound
for inhibiting expression of the at least one UCR gene, if the amount of the
UCR gene
product expressed in the cancer cells is greater than the amount of the UCR
gene product
expressed in control cells, such that proliferation of cancer cells in the
subject is inhibited.
97

42. The method of claim 41, wherein the cancer-associated chromosomal feature
is selected from the group consisting of a cancer-associated genomic region,
and a
chromosomal fragile site.
43. The method of claim 42, wherein the cancer-associated chromosomal feature
is a chromosomal fragile site.
44. The method of claim 43, wherein the cancer-associated chromosomal feature
is a cancer-associated genomic region.
45. The method of any of the preceding claims, wherein the cancer is selected
from the group consisting of: leukemia; lung cancer; esophageal cancer;
gastric cancer;
colorectal cancer; brain cancer; bladder cancer; breast cancer; cervical
cancer; epithelial
cancer; nasopharyngeal cancer; lymphoma; uterine cancer; hepatic cancer; head-
and-neck
cancer; renal cancer; male germ cell tumors; malignant mesothelioma;
myelodysplastic
syndrome; ovarian cancer; pancreatic or biliary cancer; prostate cancer;
thyroid cancer; and
urothelial cancer.
46. A marker for assessing one or more metabolic pathways that contribute to
at least
one of initiation, progression, severity, pathology, aggressiveness, grade,
activity, disability,
mortality, morbidity, disease sub-classification or other underlying
pathogenic or pathological
feature of at least one lung cancer-related disease,
wherein the marker comprises one or more UCR gene products.
47. A composition comprising one or more of the markers of the preceding
claim.
48. A method of identifying a potential for the initiation or development of
at least
one cancer-related disease in a subject, the method providing measuring one or
more of the
markers of any of the preceding claims.
49. A method of claim 48, wherein the one or more marker is present in an
isolated sample and all method steps are performed in vitro.
98

50. A reagent for testing for a cancer-related disease, wherein the reagent
comprises a polynucleotide comprising the nucleotide sequence of at least one
marker of any
of the preceding claims or a nucleotide sequence complementary to the
nucleotide sequence
of the marker.
51. A reagent for testing for a cancer-related disease, wherein the reagent
comprises an antibody that recognizes a protein encoded by at least one marker
of any of the
preceding claims.
52. A DNA chip for testing for a cancer-related disease, on which a probe has
been immobilized to assay at least one marker of any of the preceding claims.
53. A method of assessing the effectiveness of a therapy to prevent, diagnose
and/or
treat at least one lung cancer-related disease comprising:
(1) subjecting an animal to a therapy whose effectiveness is being assessed,
and
(2) determining the level of effectiveness of the treatment being tested in
treating or
preventing the lung cancer-related disease by evaluating at least one marker
of any of the preceding
claims.
54. The method of claim 53, wherein the candidate therapeutic agent comprises
one or more of: pharmaceutical compositions, nutraceutical compositions, and
homeopathic
compositions.
55. The method of claim 54, wherein the therapy being assessed is for use in a
human
subject.
56. The method of claim 55, wherein the method is not a method of treatment of
the human or animal body by surgery or therapy.
57. A method of assessing the potential of at least one material for an
ability to
initiate a cancer-related disease response in an animal model, the method
providing:
(1) measuring one or more of up- or down-regulated markers of any of the
99

preceding claims after exposure of the animal to one or more materials in
amounts sufficient to
initiate a cancer-related disease response in the animal; and
(2) determining whether at least one of the up- or down-regulated markers has
the
ability to initiate a cancer-related disease response.
58. A pharmaceutical composition for treating a cancer-related disease,
comprising:
at least one UCR gene product selected from the group consisting of a cluster
of seven
UCRs (uc.347 to uc.353);and combinations thereof; and,
a pharmaceutically-acceptable carrier.
59. The pharmaceutical composition of claim 59, wherein the at least one miR
gene product corresponds to a UCR gene product that is up- or down-regulated
in cancer cells
relative to suitable control cells.
60. The pharmaceutical composition of claim 59, wherein the lung cancer-
related
disease is adenocarcinoma.
61. A pharmaceutical composition for treating a lung cancer, comprising
at least one UCR expression-inhibition compound, and
a pharmaceutically-acceptable carrier,
wherein the at least one UCR expression-inhibition compound is specific for a
miR
gene product selected from the group consisting of a cluster of seven UCRs
(uc.347 to
uc.353); and combinations thereof.
62. The pharmaceutical composition of claim 61, wherein the at least one UCR
expression-inhibition compound is specific for a UCR gene product that is up-
or down-
regulated in cancer cells relative to suitable control cells.
63. An article of manufacture comprising: at least one capture reagent that
binds
to a marker for a cancer-related disease selected from at least one of the
markers of any of the
preceding claims.
100

64. A kit for screening for a candidate compound for a therapeutic agent to
treat a
cancer-related disease, wherein the kit comprises:
one or more reagents of at least one marker of any of the preceding claims,
and
a cell expressing at least one marker.
65. The kit of claim 64, wherein the presence of the marker is detected using
a
reagent comprising an antibody or an antibody fragment which specifically
binds with at least
one marker.
66. The kit of claim 65, wherein the reagent is labeled, radio-labeled, or
biotin-
labeled, and/or wherein the antibody or antibody fragment is radio-labeled,
chromophore-
labeled, fluorophore-labeled, or enzyme-labeled.
67. The kit of claim 66, further including a container comprising at least one
of
the markers.
68. The kit of claim 67, wherein the reagent comprises one or more of: an
antibody, a probe to which the reagent is attached or is attachable, and an
immobilized metal
chelate.
69. A screening test for a lung cancer-related disease comprising:
contacting one or more of the markers of any of the preceding claims with a
substrate
for such marker and with a test agent, and
determining whether the test agent modulates the activity of the marker.
70. A screening test of claim 69, wherein all method steps are performed in
vitro.
71. A microarray for predicting the presence of a cancer-related disease in a
subject comprising an antibody directed to at least one marker of any of the
preceding claims.
72. The method of claim 71, wherein a level of expression of the marker is
assessed by detecting the presence of a transcribed polynucleotide or portion
thereof, wherein
the transcribed polynucleotide comprises a coding region of the marker.
101

73. The method of claim 72, wherein the sample is a cancer-associated body
fluid
or tissue.
74. The method of claim 73, wherein the sample comprises cells obtained from
the patient.
75. A method for treating, preventing, reversing or limiting the severity of a
cancer-related disease complication in an individual in need thereof,
comprising:
administering to the individual an agent that interferes with at least one
cancer-related
disease response signaling pathway, in an amount sufficient to interfere with
such signaling,
wherein the agent comprises at least one UCR gene product.
76. Use of an agent that interferes with at least on lung cancer-related
disease
response signaling pathway, for the manufacture of a medicament for treating,
preventing,
reversing or limiting the severity of a cancer-related disease complication in
an individual,
wherein the agent comprises at least one UCR gene product.
77. A method of treating, preventing, reversing or limiting the severity of a
cancer-related disease complication in an individual in need thereof,
comprising
administering to the individual an agent that interferes with at least one
cancer-related disease response cascade,
wherein the agent comprises at least one UCR gene product.
78. Use of an agent that interferes with at least one cancer-related disease
response
cascade, for the manufacture of a medicament for treating, preventing,
reversing or limiting
the severity of a cancer-related disease complication in an individual,
wherein the agent comprises at least one UCR gene product.
79. A computer-readable medium comprising a database having a plurality of
digitally-encoded reference profiles, wherein at least a first reference
profile represents a
level of at least a first marker in one or more samples from one or more
subjects exhibiting an
indicia of a cancer-related disease response,
102

wherein the marker comprises one or more UCR gene products.
80. The computer readable medium of claim 79, including at least a second
reference profile that represents a level of at least a second marker in one
or more samples
from one or more subjects exhibiting indicia of a cancer-related disease
response; or subjects
having a cancer-related disease.
81. A computer system for determining whether a subject has, is predisposed to
having, or has a poor survival prognosis for, a cancer-related disease,
comprising
the database of any of the preceding claims, and
a server comprising a computer-executable code for causing the computer to
receive a profile of a subject, identify from the database a matching
reference profile that is
diagnostically relevant to the subject profile, and generate an indication of
whether the subject
has, or is predisposed to having, a cancer-related disease.
82. A computer-assisted method for evaluating the presence, absence, nature or
extent of a cancer-related disease in a subject, comprising:
(1) providing a computer comprising a model or algorithm for classifying data
from a sample obtained from the subject,
wherein the classification includes analyzing the data for the presence,
absence or
amount of at least one marker, and
wherein the marker comprises one or more miR gene products;
(2) inputting data from the biological sample obtained from the subject; and,
(3) classifying the biological sample to indicate the presence, absence,
nature or
extent of a cancer-related disease.
83. The method of claim 82, wherein the at least one UCR gene product and
combinations thereof includes isolated variants or biologically-active
fragments or functional
equivalents thereof, or antibodies that bind thereto.
84. The method of claim 83, wherein the UCR gene product is selected from the
group consisting of a cluster of seven UCRs (uc.347 to uc.353);and
combinations thereof
103

85. An animal model for cancer wherein at least one of an altered expression
of one
or more UCR gene products is present.
86. The animal model of claim 85, wherein the animal model is a nonhuman
vertebrate.
87. The animal model of claim 86, wherein the animal model is a mouse, rat,
rabbit, or primate.
104

Description

Note : Les descriptions sont présentées dans la langue officielle dans laquelle elles ont été soumises.


CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
ULTRACONSERVED REGIONS ENCODING ncRNAs
Inventor: Carlo M. Croce
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of United States Provisional
Application No.
60/963,329 filed August 3, 2007, and PCT/US2008/xxxxx filed xxxxx, 2008, the
entire
disclosure of which is expressly incorporated herein by reference.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
[0002] This invention was not made with any Government support and the
Government has
no rights in this invention.
BACKGROUND
[0003] Taken as a whole, cancers are a significant source of mortality and
morbidity in the
U.S. and throughout the world. However, cancers are a large and varied class
of diseases
with diverse etiologies. Researchers therefore have been unable to develop
treatments or
diagnostic tests which cover more than a few types of cancer.
[0004] For example, cancers are associated with many different classes of
chromosomal
features. One such class of chromosomal features are perturbations in the
genomic structure
of certain genes, such as the deletion or mutation of tumor suppressor genes.
The activation
of proto-oncogenes by gene amplification or promoter activation (e.g., by
viral integration),
epigenetic modifications (e.g., a change in DNA methylation) and chromosomal
translocations can also cause cancerigenesis. Such perturbations in the
genomic structure
which are involved in the etiology of cancers are called "cancer-associated
genomic regions"
or "CAGRs."
[0005] Chromosomal fragile sites are another class of chromosomal feature
implicated in the
etiology of cancers. Chromosomal fragile sites are regions of genomic DNA
which show an
abnormally high occurrence of gaps or breaks when DNA synthesis is perturbed
during
metaphase. These fragile sites are categorized as "rare" or "common." As their
name
suggests, rare fragile sites are uncommon. Such sites are associated with di-
or tri-nucleotide
repeats, can be induced in metaphase chromosomes by folic acid deficiency, and
segregate in
a Mendelian manner. An exemplary rare fragile site is the Fragile X site.

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[0006] Common fragile sites are revealed when cells are grown in the presence
of
aphidocolin or 5-azacytidine, which inhibit DNA polymerase. At least eighty-
nine common
fragile sites have been identified, and at least one such site is found on
every human
chromosome. Thus, while their function is poorly understood, common fragile
sites represent
a basic component of the human chromosome structure.
[0007] Induction of fragile sites in vitro leads to increased sister-chromatid
exchange and a
high rate of chromosomal deletions, amplifications and translocations, while
fragile sites
have been colocalized with chromosome breakpoints in vivo. Also, most common
fragile
sites studied in tumor cells contain large, intra-locus deletions or
translocations, and a number
of tumors have been identified with deletions in multiple fragile sites.
Chromosomal fragile
sites are therefore mechanistically involved in producing many of the
chromosomal lesions
commonly seen in cancer cells.
[0008] All malignant cells have specific alterations at DNA loci that encode
genes for
oncoproteins or tumor suppressors (Balmain et al., 2003; Wooster and Weber,
2003). This
common feature has recently been expanded to include a large class of non-
coding RNAs
(ncRNAs) called microRNAs (miRNAs) (Ambros, 2004) that are also involved in
cancer
initiation and progression (Calin et al., 2002; Croce and Calin, 2005;
Berezikov and Plasterk,
2005a; Esquela-Kerscher and Slack, 2006; Calin and Croce, 2006a). MiRNAs
affect the
regulation of gene expression at both the transcriptional and post-
transcriptional levels
(Ambros, 2003; Ambros, 2004).
[0009] The extent of involvement of miRNAs and the involvement of other
classes of
ncRNAs in human tumorigenesis is unknown. Therefore, there is a need for
further research
into the molecular mechanisms and signal transduction pathways altered in
cancer.
[00010] There is a further need for the identification of new molecular
markers and potential
therapeutic agents.
[00011] The ultraconserved regions (UCRs) of the human genome (Bejerano et
al., 2004b) are
also miRNAs that are almost completely conserved among various species
(Berezikov et al.,
2005b). For example, the active molecules of the miR-16-1/miR-15a cluster, has
been shown
to be an essential player in the initiation of chronic lymphocytic leukemia
(CLL) (Calin et al.,
2005a), and are completely conserved in human, mouse and rat and highly
conserved in nine
out of the ten sequenced primate species (Berezikov et al., 2005b).
Comparative sequence
analysis has identified a number of highly conserved genomic sequences. Some
of these
regions do not produce a transcript that is translated into protein and are
therefore considered
2

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
to be non-genic. Various names have been applied to this class of sequences:
conserved non-
genic sequences (CNGs) (Dermitzakis et al., 2005), conserved non-coding
sequences
(CNSs/CNCs) (Meisler, 2001), multiple species conserved sequences (MCSs)
(Thomas et al.,
2003) or highly conserved regions (HCRs) (Duret et al., 1993).
[00012] UCRs are a subset of conserved sequences that are located in both
intra- and inter-
genic regions. They are absolutely conserved (100%) between orthologous
regions of the
human, rat, and mouse genomes (Bejerano et al., 2004b). In contrast to other
regions of
conserved sequence, 53% of the UCRs have been classified as non-exonic ('N',
256/481
without evidence of encoding protein), while the other 47% have been
designated either
exonic ('E', 111/481, that overlap mRNA of known protein-coding genes), or
possibly exonic
('P', 114/481, with inconclusive evidence of overlap with protein coding
genes).
[00013] A large portion of transcription products of the non-coding functional
genomic
regions have significant RNA secondary structures and are components of
clusters containing
other sequences with functional non-coding significance (Bejerano et al.,
2004a). The UCRs
represent a small fraction of the human genome that are likely to be
functional but not
encoding proteins, and have been called the "dark matter" of the human genome
(Bejerano et
al., 2004a). Because of the high degree of conservation, the UCRs may have
fundamental
functional importance for the ontogeny and phylogeny of mammals and other
vertebrates.
This was illustrated by the recent finding of a distal enhancer and an
ultraconserved exon
derived from a novel retroposon active in lobe-finned fishes and terrestrial
vertebrates more
than 400 million years ago and maintained as active in a "living fossil'
coelacanth (Bejerano
et al., 2006).
[00014] Further experimental proof of the functional importance of UCRs is
based on analysis
of mice with targeted mutations. Megabase deletions of gene deserts that lack
ultraconserved
elements or highly conserved sequences resulted in viable mice that developed
apparently
without detectable phenotypes (Nobrega et al., 2004). By contrast, gene
deserts containing
several UCRs (such as the two gene deserts surrounding the DA CHI gene on
human
chromosome 13g21.33) were shown to contain long-range enhancers, some of them
composed of UCR sequences (Nobrega et al., 2003).
[00015] In spite of considerable research into therapies for cancer-related
diseases, these
diseases remain difficult to diagnose and treat effectively. Accordingly,
there is a need in the
art for improved methods for diagnosing and/or treating cancer. The present
invention fulfills
these needs and further provides other related advantages.
3

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
SUMMARY OF INVENTION
[00016] Described herein is a thorough genomic interrogation of the status of
UCRs in a large
panel of human leukemias and carcinomas.
[00017] We investigated the genome-wide expression of UCRs in various normal
and cancer
samples, and we assessed the relationship between the genomic location of
these sequences
and the known regions involved in cancers.
[00018] Furthermore, we identified a functional role for miRNAs in the
transcriptional
regulation of cancer-associated UCRs.
[00019] Also described herein is evidence in cancer systems that a
differentially expressed
UCR could alter the functional characteristics of malignant cells.
[00020] Also described herein, by combining these data with the elaborate
models involving
miRNAs in human tumorigenesis, is a model in which alteration in both coding
and non-
coding RNAs cooperate in the initiation and progression of malignancy.
[00021] In one broad aspect, there is described herein are methods for
differentiate human
cancers comprising using one or more transcribed ultraconserved regions(T-UCR)
expression
profiles where the association between the genomic location of UCRs and the
analyzed
cancer-related genomic elements is highly statistically significant and
comparable to that
reported for miRNA
[00022] Various objects and advantages of this invention will become apparent
to those skilled
in the art from the following detailed description of the preferred
embodiment, when read in
light of the accompanying drawings.
BRIEF DESCRIPTION OF THE DRAWINGS
[00023] The patent or application file contains at least one drawing executed
in color. Copies
of this patent or patent application publication with color drawing(s) will be
provided by the
Office upon request and payment of the necessary fee.
[00024] Figures 1A - 1E. Transcriptional characteristics of various types of
UCRs:
[00025] Figure 1A. Northern blots showing the expression of various UCRs in
normal
tissues. In the case of uc.246(E) and uc.269A(N), the presence of the long
transcript was
confirmed by the RACE cloning experiments. For some tissues, duplicate samples
were
procured to confirm reproducibility. Normalization was performed with U6. The
arrows on
4
SUBSTITUTE SHEET (RULE 26)

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
the left side show the identified transcripts.
[00026] Figure 1B. T-UCRs 291 and 73A expression (normalized to 18S rRNA) was
confirmed by qRT-PCR (graphs) and microarray analyses (Normalized number under
the
graph) in normal CD5+/CD19+ lymphocytes and malignant CLL samples. P-values
were
significant for both qRT-PCR and microarray data statistical comparison. Each
box
represents the distribution of expression measured for normals (blue) and CLL
patients (red),
ends of the boxes define the 25th and 75th percentiles, a line indicates the
median, bars define
the 10th and 90th percentiles.
[00027] Figure 1C. Number of UCRs expressed in one or more of 19 tissues, as
revealed by
microarray analysis; UCR type (E, N, P) numbers are indicated. Four types of
transcription
were found: ubiquitously expressed UCRs (in 18 or 19 out of 19 different
tissues), UCRs
expressed in the majority of tissues (10 to 17), UCRs expressed in a minority
of tissues (2 to
9) and tissue-specifically expressed UCRs.
[00028] Figure 1D. Percentage of each UCR type (E, N, P) that is ubiquitously
transcribed
(both uni- and bi-directionally) in all the analyzed tissues; the absolute
numbers for each
UCR type are shown in the boxes.
[00029] Figure 1E. Expression of the sense or antisense strand UCRs 73, 133
and 269,
relative to 18S rRNA, in CD19+ B cells from three different donors.
Sense/antisense strand
specific real-time RT-PCR was used to validate the strand specific expression
of the UCRs
observed with microarray analysis; the average +1- standard deviation of
microarray results
for CDS+ samples is under each graph. Microarray probes are named as follows:
the sense
genomic probe is named "+", while the probe to the complementary sequence is
named "A+".
[00030] Figures 2A-2B. Hierarchical clustering of tissues and tumors according
to UCRs
expression. Unsupervised cluster of (Figure 2A) 22 normal human tissues and
(Figure
2A\B) 133 leukemias and carcinomas made using the non-exonic UCRs of the chip.
Some of
the T-UCRs that well differentiate the tissue types (Figure 2A) or carcinomas
from
leukemias (Figure 2B) are expanded at the right. Samples are in columns, T-
UCRs in rows.
A green colored gene is down-regulated compared to its median expression in
all samples,
red is up-regulated and yellow means no variation. The complete UCRs profile
of tissues and
tumors can be found in Figures 5 and 6.
[00031] Figures3A-3E. T-UCRs represent direct targets of miRNAs (SEQ ID Nos.1-
3, 2, 4-8
SUBSTITUTE SHEET (RULE 26)

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
and 2, respectively, in order of appearance):
[00032] Figure 3A. Examples of sites of complementarity T-UCR::miRNA. The
uc.348::
miR-155 pairing is shown as an example of low levels of complementarity in
contrast with
the other 4 interacting paired genes for which higher levels of
complementarity are found.
[00033] Figure 3B. The correlation by qRT-PCR for miR-155, uc.160 and uc.346A
expression in 9 CLL patients. Lymphocytes from four different individuals were
used as
normal controls.
[00034] Figure 3C. The direct miRNA::T-UCR interaction. Relative repression of
firefly
luciferase expression standardized to a transfection control, Renilla
luciferase. pGL-3
(Promega) was used as the empty vector. All the experiments were performed
four to eight
times in triplicate (n=12-24).
[00035] Figure 3D. The effects of miR-155 transfection in MEG-O1 cells on
expression
levels of uc.160 and uc.346A. Effects were measured by qRT-PCR at 0, 24 and 48
hours
post-transfection.
[00036] Figure 3E. Two scatter plots between expression values of mir-24-1 and
uc.160 and
of miR-155 and uc.346A are presented. The regression line shows the negative
correlation
between these two genes. The name of the corresponding array probes are
presented on the
Y and X axes. Both probes recognize the mature form of the miRNA gene.
[00037] Figures 4A-4D. T-UCR 73A(P) is acting as an oncogene in colon cancer
cells:
[00038] Figure 4A. The expression inhibition by various siRNAs in COLO-320
cells. As
reference value we used a siRNA control from Dharmacon. The most effective two
siRNAs
and a pool of four different siRNAs, including these two, were used.
[00039] Figure 2B. The antiproliferative effects of reduction in uc.73A(P)
gene expression
using siRNA-uc73A in COLO-320 colorectal cancer cells. All the results
represent the
median of three independent triplicate experiments. The levels of uc. 73A (P)
expression
were measured by RT-PCR. Two asterisks indicate a statistically significant
effect at P<0.01,
while one at P<0.05.
[00040] Figure 4C. Reduced levels of uc.73A(P) (using various siRNAs) results
in enhanced
apoptosis as shown by the Annexin-V staining assay in COLO-329 cells. As
reference value
we used a siRNA control from Dharmacon.
6
SUBSTITUTE SHEET (RULE 26)

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[00042] Figure 4D. Inhibition of uc.73A(P) by various siRNAs did not influence
SW620
colon cancer cell survival. All the results represent the median of three
independent triplicate
experiments.
[00043] Figure 5. UCR expression in human normal and malignant tissues by
Northern blot.
The expression for uc.192(N) and uc.246(E) in normal mononuclear cells (MNC)
and CLL
samples is presented. Normalization was done with U6 probe. The arrows on the
left side
show the identified transcripts. Under the gel image there are the averages of
UCR
normalized expression values in CLL and MNC samples from microarray
experiments; p-
values were from ANOVA statistic.
[00044] Figure 6. UCR expression in human normal and malignant tissues by qRT-
PCR.
Relative gene expression by qRT-PCR in CD5+/CD19+ positive lymphocytes and
human
chronic lymphocytic leukemia (CLL) samples. UCR microarray values of CLL and
CD5+
samples are indicated under the graph; p-values were from ANOVA statistic.
[00045] Figure 7. T-UCR expression profile of 22 normal human tissues.
Clustering of
tissues and UCRs revealed a distinct pattern of UCRs expression in normal
human tissues.
Samples are shown in columns, T-UCRs in rows. A green colored gene is down-
regulated
compared to its median expression in all samples, red is up-regulated and
yellow means no
variation.
[00046] Figure 8. T-UCR expression profile of 173 cancers and corresponding
normal
tissues. Samples from leukemia and normal blood cells are separated from
cancer and tissue
of epithelial origin. Samples are shown in columns, T-UCRs in rows. A green
colored gene
is down-regulated compared to its median expression in all samples, red is up-
regulated and
yellow means no variation.
[00047] Figure 9. The expression of uc.73A(P) gene in various colon cancer
cell lines by
quantitative RT-PCR. The expression in normal colon represents the median
value of 4
different samples. For normalization we used beta-actin.
[00048] Figure 10. The uc.73A(P) inhibition by siRNA1 in COLO-320 and SW-620
cells at
48hrs. Comparable levels of inhibition in respect with a siRNA of control
(Dharmacom)
were achieved in both types of cells. In spite of this, the biological effects
were seen only in
COLO-320 cells, where the T-UCR is overexpressed about 2.5 times when compared
with
expression in normal colon.
7

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[00049] Figure 11. Downregulation by small interfering of uc.73A(P) induces
apoptosis in
COLO-320 cells but not in SW-620 cells. Data obtained with caspase-3 assay in
COLO-320
(upper panel), where a significant increase in apoptotic cells is found, and
in the control
cells SW620 (lower panel), where no difference could be found. COLO-320
express high
levels of uc.73A(P), while in SW620 the expression is comparable with the
normal colon
levels.
[00050] Figure 12 - Table 1. Most significant differentially expressed UCRs in
leukemias
and carcinomas.
[00051] Figure 13 - Table 2. Mixed effect Poisson regression results as
association of UCRs
with regions of interest.
[00052] Figure 14 - Table 3. T-UCRs whose expression inversely correlates with
complementary miRNA differentially expressed in CLL patients.
[00053] Figure 15 - Table 4. T-UCRs expression in 22 human normal tissues (3
in duplicate,
from 2 different individuals).
[00054] Figure 16 - Table 5. T-UCRs differentially expressed in CLL, CRC and
HCC
identified by ANOVA analysis at P<0.005 (GeneSpring GX software).
[00055] Figure 17 - Table 6. Genomic location of UCRs is correlated with CAGR.
(databases as in (Bejerano et al., 2004); (Calin et al., 2004)).
[00056] Figure 18 - Table 7. Negative correlations between the expression of
miRNAs and
T-UCRs in CLL patients. All validated negative correlation by FDR method at
0.01
threshold, or 1% of false positive results, and with an R correlation lower as
0.40 were
considered.
DESCRIPTION OF EMBODIMENTS
[00057] As used herein, a "CAGR" includes any region of the genomic DNA that
comprises a
genetic or epigenetic change (or the potential for a genetic or epigenetic
change) that differs
from normal DNA, and which is correlated with a cancer. Exemplary genetic
changes
include single- and double-stranded breaks (including common breakpoint
regions in or near
possible oncogenes or tumor-suppressor genes); chromosomal translocations;
mutations,
deletions, insertions (including viral, plasmid or transposon integrations)
and amplifications
8

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
(including gene duplications) in the DNA; minimal regions of loss-of-
heterozygosity (LOH)
suggestive of the presence of tumor-suppressor genes; and minimal regions of
amplification
suggestive of the presence of oncogenes. Exemplary epigenetic changes include
any changes
in DNA methylation patterns (e.g., DNA hyper- or hypo-methylation, especially
in promoter
regions).
[00058] Many of the known miR genes in the human genome are in or near CAGRs,
including
80 miR genes that are located exactly in minimal regions of LOH or minimal
regions of
amplification correlated to a variety of cancers. Other miR genes are located
in or near
breakpoint regions, deleted areas, or regions of amplification.
[00059] For example, cancers associated with CAGRs include leukemia (e.g.,
AML, CLL,
pro-lymphocytic leukemia), lung cancer (e.g., small cell and non-small cell
lung carcinoma),
esophageal cancer, gastric cancer, colorectal cancer, brain cancer (e.g.,
astrocytoma, glioma,
glioblastoma, medulloblastoma, meningioma, neuroblastoma), bladder cancer,
breast cancer,
cervical cancer, epithelial cancer, nasopharyngeal cancer (e.g., oral or
laryngeal squamous
cell carcinoma), lymphoma (e.g., follicular lymphoma), uterine cancer (e.g.,
malignant
fibrous histiocytoma), hepatic cancer (e.g., hepatocellular carcinoma), head-
and-neck cancer
(e.g., head-and-neck squamous cell carcinoma), renal cancer, male germ cell
tumors,
malignant mesothelioma, myelodysplastic syndrome, ovarian cancer, pancreatic
or biliary
cancer, prostate cancer, thyroid cancer (e.g., sporadic follicular thyroid
tumors), and
urothelial cancer.
[00060] As used herein, a "FRA" includes any rare or common fragile site in a
chromosome;
e.g., one that can be induced by subjecting a cell to stress during DNA
replication. For
example, a rare FRA can be induced by subjecting the cell to folic acid
deficiency during
DNA replication. A common FRA can be induced by treating the cell with
aphidocolin or 5-
azacytidine during DNA replication. The identification or induction of
chromosomal fragile
sites is within the skill in the art; see, e.g., Arlt et al. (2003),
Cytogenet. Genome Res. 100:92-
100 and Arlt et al. (2002), Genes, Chromosomes and Cancer 33:82-92, the entire
disclosures
of which are herein incorporated by reference.
[00061] Approximately 20% of the known human miR genes are located in (13
miRs) or
within 3 Mb (22 miRs) of cloned FRAs. Indeed, the relative incidence of miR
genes inside
fragile sites occurs at a rate 9.12 times higher than in non-fragile sites.
Moreover, after
studying 113 fragile sites in a human karyotype, it was found that 61 miR
genes are located in
9

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
the same chromosomal band as a FRA.
[00062] For example, cancers associated with FRAs include bladder cancer,
esophageal
cancer, lung cancer, stomach cancer, kidney cancer, cervical cancer, ovarian
cancer, breast
cancer, lymphoma, Ewing sarcoma, hematopoietic tumors, solid tumors and
leukemia.
[00063] The following examples are included to demonstrate preferred
embodiments of the
invention. It should be appreciated by those of skill in the art that the
techniques disclosed in
the examples which follow represent techniques discovered by the inventor to
function well
in the practice of the invention, and thus can be considered to constitute
preferred modes for
its practice. However, those of skill in the art should, in light of the
present disclosure,
appreciate that many changes can be made in the specific embodiments which are
disclosed
and still obtain a like or similar result without departing from the spirit
and scope of the
invention.
[00064] EXAMPLE 1
[00065] Genome-wide profiling reveals extensive transcription of
ultraconserved regions
(UCRs) in normal human tissues.
[00066] To investigate the involvement of UCRs in human cancers, we analyzed
481 genomic
regions longer than 200 bp (Bejerano et al., 2004b) by Northern blot,
quantitative PCR (qRT-
PCR) and microarray.
[00067] Both exonic (E) and non-exonic (N) UCR probes detected transcripts (in
sense or
antisense - A, orientation) over a large range of lengths from various normal
tissues (Figure
1A and Figure 5).
[00068] The length of two of the transcripts was confirmed by cloning the cDNA
by 5'- and 3'-
RACE for the exonic uc.246(E) from normal human colon and the non-exonic
uc.269A(N)
from normal human bone marrow. Neither of these cDNAs contained open reading
frames
(ORFs) of significant length, confirming their likely non-protein coding
nature. These non-
spliced full-length cDNAs, that we named non-coding ultraconserved genes, nc-
UCGs, are of
variable length (about 0.8 kb for the ultraconserved gene UCG.246 and about
1.8kb and
2.8kb for the ultraconserved gene UCG.269A).
[00069] Transcription of these nc-UCGs may be initiated from poly-adenine rich
genomic
regions, as was recently proposed for several long ncRNAs from mouse (Furuno
et al., 2006).

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[00070] We compared the transcription levels of several UCRs from normal and
disease tissue
using microarray analysis followed by qRT-PCR and Northern blot confirmation.
The
expression of uc.291(P) and uc. 73A(P) was significantly higher in normal
CD5+/CD 19+
lymphocytes than in CLL cells (P < 0.05) (Figure 1B). The data obtained with
this
microarray platform has been confirmed in various studies (Calin et al.,
2005a; Yanaihara et
al., 2006; Volinia et al., 2006).
[00071] The strength of our data is reinforced by the fact that two
independent sets of normal
CD5 cells were included in microarray and quantitative RT-PCR experiments.
When both
uc.291(P) and uc.73A(P) were investigated by qRT-PCR and microarray in two
different sets
of CD5/CD19 positive B cells and malignant B cells, the differential
expression was
statistically significant by both assays (Figure 1B).
[00072] Furthermore, qRT-PCR and Northern blotting for eleven and six UCRs,
respectively,
gave results that were concordant with microarray results (Figure 5 and Figure
6).
[00073] Using microarray analysis, we found that the majority of transcribed
UCRs (that we
named here T-UCRs) were expressed in normal human tissues both ubiquitously
and in a
tissue-specific manner (Figure 1C).
[00074] About 34% of putative T-UCRs (325/962) had hybridization signals with
an intensity
over background (calculated as average signal of blank spots + 2 SD) in all 19
tissue samples.
The highest number of T-UCRs was found in B cells, while the lowest was in
ovary. About
93% of the UCRs (890 of 962) were expressed over background in at least one
sample, and
therefore we considered these as T-UCRs. The three different types of UCRs
were
transcribed with similar frequencies: 41% of exonic UCRs, 33% of possibly
exonic UCRs
and 30% of non-exonic UCRs.
[00075] The microarray platform contains putative T-UCRs in both sense and
anti-sense
orientation. Eighty-four of the 962 UCRs (9%) were bidirectionally
transcribed, while 241
were transcribed only from one strand, in all the normal tissues analyzed
(Figure 1D, Figure
1E and Table 4).
[00076] Since identification of bidirectional transcription by microarray
analysis may be
hindered by trace contamination with genomic DNA, we performed a comparison of
microarray results with strand-specific qRT-PCR for uc.2 69(N), uc.233(E) and
uc. 73(P). In
all three instances the data were concordant, showing predominant
transcription from one
11

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
strand (Figure 1E).
[00077] Of note, out of the 156 non-exonic T-UCRs expressed in all 19 tissues,
92 (-60%) are
intergenic, while 64 are intronic. Of the latter, 37 are in the antisense
orientation compared
with the host gene, suggesting that about 83% (129/156) of the non-exonic T-
UCRs did not
represent intronic transcription of long precursor transcripts of known host
genes, but bona
fide independent noncoding transcripts.
[00078] As with miRNAs (Liu et al., 2004), we performed a hierarchical
clustering of T-UCR
expression in hematopoietic tissues (represented by B lymphocytes, T
lymphocytes and
mononuclear cells, each collected from two healthy individuals) and non-
hematopoietic
tissues. The same types of tissue from different individuals were clustered as
closest
neighbors (Figure 2A and Figure 7).
[00079] These findings demonstrate that UCRs represent, in a significant
proportion of cases,
non-coding transcripts in normal human tissues and that the expression of
these T-UCRs is
tissue-specific.
[00080] Distinct UCR signatures in human leukemias and carcinomas
[00081] Since extensive gene expression alterations in cancer cells have been
widely
described for both protein coding genes and miRNAs (Esquela-Kerscher and
Slack, 2006;
Calin and Croce, 2006a; Calin and Croce, 2006b; Lu et al., 2005), we
investigated the
expression of UCRs in a panel of 173 samples, including 133 human cancers and
40
corresponding normal tissues.
[00082] Hierarchical clustering of the samples showed that various types of
cancers clustered
differently according to their developmental origins: the leukemias (CLL) and
normal
hematopoietic tissues were branched separately from the colorectal (CRC) and
hepatocellular
carcinomas (HCC) with their normal counterparts (Figure 8); moreover, specific
groups of
UCRs seemed to be differentially expressed in tumor types (Figure 2B).
[00083] Since different tissues have specific UCR signatures, this clustering
pattern could be
the consequence of different tissue-specific origin of the tumors. Thus, we
compared the
expression of UCRs between the normal and tumor cells of the same origin. Out
of 962
possible T-UCRs, 88 (9.1%) were differentially expressed at a highly
statistically significant
level (P<0.005) in at least one type of cancer (Table 1 and Table 5).
12

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[00084] We found both down-regulated and up-regulated T-UCRs in cancers
compared to the
expression in corresponding normal tissues. By comparing each cancer type with
the
corresponding normal tissues, we found that the CLL signature was composed of
19 UCRs (8
up- and 11 down-regulated), the CRC signature of 61 UCRs (59 up- and 2 down-
regulated),
and the HCC signature of 8 UCRs (3 up- and 5 down-regulated) (Table 5).
[00085] Eighteen transcripts of the signatures were exonic UCRs (20%), 28 were
possibly
exonic UCRs (32%) and 42 were non-exonic UCRs (48%). Of the 18 exonic T-UCRs,
9
represented the anti-sense direction of the known host protein-coding gene
transcripts. We
therefore demonstrated that the T-UCR expression profiles can be used to
differentiate human
cancers.
[00086] UCRs are frequently located at fragile sites and genomic regions
involved in
cancers
[00087] We compared the genomic location of UCRs with that of previously
reported non-
random genetic alterations identified in human tumors and cloned fragile sites
(FRA) as
described (Calin et al., 2004b). We used the set of 186 miRNAs previously
reported (Calin et
al., 2004b) and a set of 297 zinc finger protein-coding genes (ZNF)
(genome.ucsc.edu), a
well known family of transcription factors shown to be associated with cancer
(Huntley et al.,
2006).
[00088] We previously reported that miRNA genes are frequently located at FRA
sites, HOX
genes clusters and genomic regions involved in cancer, such as minimal regions
of loss of
heterozygosity (LOH), and minimal regions of amplification, globally named
cancer
associated genomic regions (CAGR) (Calin et al., 2004b).
[00089] A recent study, using high-resolution array comparative genomic
hybridization
(aCGH), confirmed that miRNA loci exhibit genomic alterations at high
frequency in human
cancers (Zhang and al, 2006). Furthermore, by analyzing the miRNA expression
in NCI-60
cell lines, another group found that the candidate tumor-suppressor and
oncogenic miRNAs
are located in CAGRs (Gaur et al, 2007).
[00090] Here, we show that the association between the genomic location of
UCRs and the
analyzed cancer-related genomic elements is highly statistically significant
and comparable to
that reported for miRNAs. The ZNF transcription factors did not show any
significant
association with any of the analyzed regions of interest (Table 2 and Table
6).
13

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[00091] There was a similar lack of association for the smaller family of
protein-coding genes
involved in RNA splicing (80 genes, data not shown). For example, the
probability for the
association of UCRs or miRNAs with minimal LOH regions versus non-deleted
genomic
regions was less than 0.001 in both instances (IRR of 2.02 and 4.08,
respectively). As an
internal control, we used the human papilloma virus 16 (HPV 16) integration
sites, which
frequently occur in FRA sites. If UCRs are significantly associated with FRA,
then we
expected to fin an association with the HPV 16 integration site. This is
exactly what we
observed for both UCRs and miRNAs, but not for ZNF protein-coding genes (Table
2) or for
the protein-coding genes involved in RNA splicing (data not shown).
[00092] Additional data illustrate the importance of the genomic location of
UCRs. First, we
found that the ubiquitously expressed T-UCRs (expressed in 18 or 19 normal
tissues in
Figure 1 C) are significantly more frequently located in CAGRs (P<0.005,
Fisher exact test)
when compared with all other UCRs (97 out of 189 vs. 71 out of 292). Second, T-
UCRs
differentially expressed in human cancers are located in CAGRs specifically
associated with
that type of cancer. For example, the chromosomal region 13g21.33-g22.2 has
been linked to
susceptibility to familial CLL (Ng et al, 2007). No mutations were found in
any of the 13
protein-coding genes screened within this interval.
[00093] We identified a cluster of seven UCRs (uc.347 to uc.353) located
within this CAGR.
Two of them, uc.349A(P) and uc.352(N) are among the T-UCRs that are
differentially
expressed between normal and malignant B-CLL CD5 positive cells.
[00094] This suggests, at least in this case, that it is not the protein-
coding genes but the UCRs
that represent the "unknown" culprits located in the CAGR. Together these data
provide
evidence that the UCRs are located in genomic regions altered during the
malignant process,
and suggest that T-UCRs could be candidate genes for cancer susceptibility.
[00095] Negative regulation of T-UCRs by direct interaction with microRNAs
[00096] In order to begin to functionally characterize some UCRs involved in
human cancers,
we performed a genome-wide expression study in the same set of CLL samples
investigated
above. We found that a signature of five UCRs, uc.269A(N), uc.160(N),
uc.215(N),
uc.346A(P) and uc.348(N), was able to differentiate between two main CLL
prognosis
groups previously differentiated by the expression of 70-kDa zeta-associated
protein (ZAP-
70).
14

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[00097] These five T-UCRs displayed variations in their expression level that
was negatively
correlated with the miRNA expression signature reported in CLL (Cahn et al.,
2005a) (Table
3).
[00098] While not wishing to be bound by theory, the inventor herein now
believes that this
finding raises the possibility of complex regulatory mechanisms between miRNAs
and T-
UCRs. We identified, by sequence alignments, that three out of the 5 UCRs have
significant
antisense complementarity with 5 out of the 13 miRNAs from the signature,
giving rise to six
possible interacting pairs: uc.160:: rniR-24, uc.160:: miR-155, uc.160:: miR-
223,
uc.160::miR-146a, uc.346A::miR-155, and uc.-348::miR-29b (Figure 3A).
[00099] In this analyzed set of miRNA::UCR pairs, the 5'- end "6 base seed"
complementarity
rule described for miRNA::mRNA interaction was valid; furthermore, the levels
of 3 '-end
complementarity could be variable: more than 60 Io complementarity for miR-
24:: uc160 or
miR-155::uc.346A pairs to less than 25% for the miR-155:: uc.160 pair. As a
control, when
randomly generated combinations of five UCRs and 13 miRNAs were compared, the
sense
and antisense complementarity was not significant.
[000100] Negative correlations between the microa7ay expression values of
specific T-UCRs
and predicted interactor miRNAs was confirmed by qRT-PCR for selected T-UCRs
and
miRNAs from lymphocytes of an independent set of CLL patients and normal
controls
(Figure 3B).
[000101] We performed in vitro assays of miRNA::UCR interaction involving miR-
155 which
is overexpressed in the aggressive form of CLL (Calin et al., 2005) some
lymphomas and
carcinomas (Eis et al., 2005; Kluiver et al., 2005; Volinia et al., 2006), and
miR-24-1 and
miR29-b which carry mutations in primary transcripts from CLL patients (Calin
et al.,
2005a).
[000102] We cloned the UCRs uc.160(N), uc.346A(P) and uc.348(N) in luciferase
reporter
vectors to assess the possible direct interaction with miR-155, miR-24-1 or
miR-29-b. We
observed consistent and reproducible reduction in luciferase expression with
four miR::T-
UCR pairings consistent with interactions taking place in vitro (Figure 3C).
[000103] By contrast, uc.348(N) did not interact with miR155 as indicated by
the luciferase
assay, a result that is in concordance with the positive expression
correlation of these two
genes in CLL patients and the low sequence complementarity (Figure 3A).

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000104] Interactions in vivo
[000105] In order to determine if these interactions occur in vivo, we
transfected miR-155 into
MEG01 leukemia cells and assessed the levels of uc.346A and uc.160 (both well
expressed in
this cell line). At 24 hours after transfection, miR-155 significantly reduced
the expression
level of both T-UCRs; after 48 hours, the reduction of exogenous miR-155
levels was
paralleled by an increase in T-UCR expression (Figure 3D).
[000106] This reversible effect supports a regulation of T-UCR by specific
miRNAs. As this
interaction was proven for the genes of the "ZAP-70 signature", we
investigated the
correlations between the expression of all miRNAs and T-UCRs at the genome-
wide level in
all 50 CLL patients. Interestingly, we found a significant negative
correlation (at a false
detection rate (FDR) of less than 0.01) between 87 miRNAs (out of 235 spotted
on the chip,
37%) and T-UCRs expression levels (Table 7).
[000107] Furthermore, among the correlated genes we identified the miR-24-1::
uc.160 and the
miR155:: uc346A(P) pairs, experimentally proven to interact (Figure 3E).
[000108] Moreover, miR-155 and uc.348, that did not interact experimentally,
were not
members of this list. Other pairs of possible interactors for which we
identified positive
luciferase assays were miR-15-a:: uc.78 and miR16:: uc. 78 (data not shown).
Therefore,
non-coding T-UCRs represent possible targets of miRNAs, and these interactions
may have
biological and prognostic significance for cancer patients.
[000109] T-UCRs may act as Oncogenes
[000110] To expand the functional characterization of T-UCR, we examined the
biological
effects of uc. 73A (P) in a cancer model. Since this is one of the most
statistically significant
up-regulated T-UCRs in colon cancers (P < 0.001), we decided to investigate
the effects of its
downregulation in COLO-320 colorectal cancer cells that expressed high levels
of uc.73A(P).
As a control we used the SW620 colon cancer cells in which the expression of
this gene does
not differ from normal colonic cells (Figure 9).
[000111] Two small interfering RNAs (siRNA1 and siRNA3), as well as a pool of
four siRNAs
(siRNApool), were designed to target uc. 73A(P) and transfected into COLO-320
and SW620
cells. There was significantly less expression of uc.73A(P) after 48 (Figure
4A and Figure
10), 72 and 144 hours (data not shown) in the COLO-320 cells treated with
siRNAs 1, 3 and
pool. The same was found also for SW-620 cells (Figure 6).
16

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000112] The growth of COLO-320 cells was significantly reduced after 144
hours of treatment
with specific siRNA compared to both untreated (null) or siRNA-treated control
cells (P <
0.05 at 96 hrs and P < 0.005 at 144hrs) (Figure 4B).
[000113] In comparison, proliferation of the SW620 control cells was not
significantly changed
(P = 0.83 and P = 0.23 at 96 and 144 hrs, respectively) (data not shown). Cell
cycle studies
revealed an increase in the sub-Gl fraction of cells (suggesting the presence
of apoptotic cells,
data not shown) in COLO-320 cells, but not in SW620 cells, a finding confirmed
by the
apoptosis-specific AnnexinV assay (Figure 4C and Figure 4D) and by caspase-3
assay
(Figure 11).
[000114] Furthermore, the intensity of effects on cell proliferation and
survival were
proportional with the degree of inhibition by siRNAs (Figure 4).
[000115] These data suggest that in colorectal cancers, uc. 73A(P) behaves
like an oncogene by
increasing the number of malignant cells as a consequence of reduced
apoptosis.
[000116] DISCUSSION
[000117] According to the dogma of molecular oncology, cancer is a genetic
disease involving
tumor-suppressor and oncogenic proteins (Bishop, 1991; Hunter, 1991; Weinberg,
1991).
Recent findings strongly support the involvement of microRNAs in the
pathogenesis of a
majority of analyzed cancers, and add a new layer of complexity to the
molecular architecture
of human cancers (Calin et al., 2002; Esquela-Kerscher and Slack, 2006; Calin
and Croce,
2006a). MiRNAs represent, however, just a particular group of ncRNAs involved
in human
cancers. It has been shown that antisense intronic ncRNA levels correlate with
the degree of
tumor differentiation in prostate cancer (Reis et al., 2005) and that MALAT-1
ncRNA
expression predicts metastasis and survival in early stage non-small cell lung
cancer (Ji et al.,
2003), suggesting a deeper link between ncRNAs and tumor biology.
[000118] To clearly address this question, we investigated at the genomic
level a full new class
of ncRNAs, namely the transcribed non-coding ultraconserved regions (T-UCRs).
We used
bioinformatics tools to demonstrate that the UCRs are located in genomic
regions targeted
during the malignant process indicative of a putative involvement in human
tumorigenesis.
[000119] As now shown herein, we were able to clone by RACE amplification
cDNAs
corresponding to uc.246(E) and uc.269A(N), proving that the UCRs are bona fide
genes
(named herein as nc-UCGs) that are expressed and can be cloned by standard
methods.
17

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000120] Various expression techniques including Northern blot, qRT-PCR and
genome-wide
microarray profiling, proved that UCRs are frequently transcribed and that
there are distinct
signatures in human leukemias and carcinomas. We focused on chronic
lymphocytic
leukemia, the most frequent adult leukemia in the Western world (Chiorazzi et
al., 2005), on
colorectal carcinoma, one of the most common cancers in industrialized
countries (de la
Chapelle, 2004) and on hepatocellular carcinoma, the most rapidly increasing
type of cancer
in America (Wilson, 2005).
[000121] We found that for all the tumor types examined, the malignant cells
have a unique
spectrum of expressed UCRs when compared with the corresponding normal cells,
suggesting that significant variations in T-UCR expression are involved in the
malignant
process.
[000122] Characterizing the functional significance of T-UCR alterations in
human cancers is
not a trivial task. A myriad of putative functions of T-UCRs can be
hypothesized, including
an antisense inhibitory role for protein coding genes or other non-codingRNAs,
or a role as
"aspecific" miRNAs, meaning miRNAs with peculiarities such as very long
precursors (e.g.
uc.339(P) which has a precursor length that is double the usual miRNA). This
puzzle
becomes more complicated by the fact that several UCRs do not act like genes
and were
found to have regulatory functions as enhancers (Nobrega et al., 2003;
Pennacchio et al.,
2006), while others represent exons of protein coding genes with known/unknown
cancer
connections. A particularly interesting region is the DA CHI locus that
contains 7 UCRs in
about 700kb (Bejerano et al., 2004b). Three of the UCRs from this region are
differentially
expressed in analyzed cancers, two of which are members of the CLL signature.
The
majority of scanned conserved regions from this locus in a mouse model are
enhancers,
including the uc.351(N) that was not expressed in any of the analyzed tissues
in our study.
[000123] Interestingly, the only two regions that failed to have enhancer
function are uc.348(N)
and uc.352(N), both classified as non-coding and both differentially expressed
in human
cancers. Further increasing the interest in these specific T-UCRs, is the
finding that this
genomic region has been linked to susceptibility to familial CLL and that none
of the known
protein-coding genes were mutated (Ng et al, 2007).
[000124] Recently, it was found that short blocks of several tens of bp from
the noncoding parts
of the human genome (named pyknons) occur within nearly all known protein
coding genes
(Rigoutsos et al., 2006). While the pyknons are distinct from the UCRs, the
ultraconserved
18

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
element containing the highest number of pyknons (four) was uc. 73(P), which
we found to
be one of the most differentially expressed T-UCRs in both CLL and CRC. These
intriguing
observations suggest a possible regulatory role for uc. 73(P) on the coding
genes with
complementary sequences.
[000125] Further expanding the involvement of this T-UCR in human cancers, we
were able to
prove an oncogenic function for uc.73(P) in colon cancer, as diminution of its
over-
expression induced apoptosis and had antiproliferative effects specifically in
colon cancer
cells abnormally expressing this T-UCR.
[000126] Our findings that another class of ncRNAs, the T-UCRs, is
consistently altered at the
genomic level in a high percentage of analyzed leukemias and carcinomas,
support a model
in which both coding and non-coding genes are involved and cooperate in human
tumorigenesis (Calin and Croce, 2006b).
[000127] Furthermore, correlations between the expression of UCRs and miRNAs
in CLL
patients raise the intriguing possibility of complex functional regulatory
pathways in which
two or more types of ncRNAs interact and influence the phenotype.
[000128] We also demonstrated the existence of the miRNA::T-UCR interaction in
which two
different types of ncRNAs are interacting..
[000129] We found that nc-UCGs are consistently altered at the genomic level
in a high
percentage of leukemias and carcinomas, and may interact with miRNAs in
leukemias. The
findings provide support for a model in which both coding and non-coding genes
are
involved in and cooperate in human tumorigenesis.
[000130] EXAMPLE I
[0001311 EXPERIMENTAL PROCEDURES
[000132] A) RACE cloning and expression analysis by microarray, qRT-PCR and
Northern
blot
[000133] 1) RACE cloning
[000134] The expression of six UCRs (uc.47(N), uc.110(N), uc.192(N),
uc.246(E), uc.269A(N)
and uc.352(N)) was analyzed in brain, testis, bone marrow, small intestine,
colon and liver
tissue using various combinations of PCR primers designed to amplify short
products. These
products included 40-mers used for probes in microarray analysis and the
complete >200bp
19

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
UCR sequence. Two of the UCR products, one exonic, uc.246(E) and one non-
exonic,
uc.269A(N), were cloned by Rapid Amplification of cDNA Ends (RACE) in both 5'
and 3'
directions. The sources of tissue from which sequences were cloned were bone
marrow,
leukocytes, fetal brain and colon according to the manufacturer protocol
(Marathon-ready
cDNAs, Clontech, Palo Alto, CA).
[000135] 2) UCR expression study by microarray.
[000136] Total RNA was extracted with Trizol (Invitrogen, Carlsbad, CA) from
19 normal
human tissues (Liu et al., 2004) and from 50 CLL samples from patients
diagnosed with
CLL. Informed consent was obtained from all patients at the CLL Research
Consortium
institutions in the US. As controls, CDS+ B cells from 6 healthy individuals
(four distinct
samples, two being pools from two different healthy individuals) and
mononuclear cells
(MNC) from 3 individuals were used as reported in (Calin et al., 2005a). RNA
was also
extracted from 78 primary colorectal carcinomas, 21 normal colonic mucosas, 9
primary
hepatocellular carcinomas and 4 normal livers, collected at the University of
Ferrara,
University of Bologna and University Tor Vergata, Rome (Italy). All samples
were obtained
with written informed consent according to institutional guidelines for the
protection of
human subjects.
[000137] Microarray chips were developed with a total of 481 human UCR
sequences as in
(soe.ucsc.edu/-jill/ultra). For each UCR two 40-mer probes were designed, one
corresponding to the sense genomic sequence (named "+") and the other to the
complementary sequence (named "A+"). The design criteria were as described
(Liu et al.,
2004). Each oligo was printed in duplicate in two different slide locations,
and therefore
quadruplicate numerical values were available for analysis. Several thousand
(3484) blank
spots were used for background subtraction. RNA extraction and microarray
experiments,
consisting of the UCR microarray assembly, target preparation and array
hybridization, were
performed as described in detail elsewhere (Liu et al., 2004; Calin et al.,
2004a).
[000138] Briefly, 5 g of RNA from each tissue sample was labeled with biotin
by reverse
transcription using random hexamers. Hybridization was carried out on the
second version of
our miRNA-chip (ArrayExpress accession number: A-MEXP-258) which contained the
962
UCR probes, 238 probes for mature miRNA and 143 probes for precursor miRNAs.
Each
oligo was printed in duplicate in two different slide locations. Hybridization
signals were
detected by biotin binding of a Streptavidin - A1exa647 conjugate (one-color
signal) using a

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
GenePix 4000B scanner (Axon Instruments) . Images were quantified using the
GenePix Pro
6.0 (Axon Instruments).
[000139] Raw data were normalized and analyzed in GeneSpring GX 7.3 (Agilent
Technologies, Santa Clara, CA). Expression data of the 22 tissue samples were
normalized
with Lowess function in Bioconductor (Limma package) and then were median
centered
using GeneSpring normalization; the threshold used to determine the level of
UCR
expression was calculated as the average of blank spots + 2 SD (standard
deviation). Tumors
were normalized using the on-chip and on-gene median normalization of the
GeneSpring
software. Hierarchical cluster analysis was done using average linkage and
Pearson
correlation as measures of similarity. Statistical comparisons of tumors and
normal tissues
were performed by filtering on fold change and then using the ANOVA (Analysis
of
Variance) statistic of the GeneSpring software and the Benjamin and Hochberg
correction for
reduction of false positives. The filter on fold-change was set on 1.2 because
this threshold,
already used for microRNAs analyzed with the same chip [see for examples (Cahn
et al.,
2005a; Cimmino et al., 2005; Iorio et al., 2005)], was demonstrated to reflect
a real biological
difference. The T-UCRs differentially expressed among CLL patients, grouped in
accordance to 70-kDa zeta-associated protein (ZAP-70) expression, were
identified by
combining the ANOVA results with the SAM (Significance Analysis of Microarray)
and
PAM (Prediction Analysis of Microarrays) analysis. Their expression was
compared to that
of microRNAs (Calin et al., 2005a). All data were submitted using MIAMExpress
to the
ArrayExpress database and could be retrieve using the accession number E-TABM-
184.
[000140] 3) Quantitative RT-PCR for UCRs.
[000141] Quantitative RT-PCR was the first method we used to confirm the
microarray results.
We validated the microarray data for eleven UCRs, including uc. 73 (P)/73A (P)
, uc.135(E),
uc. 160(N), uc.233(E)/233A(E), uc.269(N)/269A(N), uc.289(N), uc.291(P), and
uc.346A(P)
in various combinations of samples, including 15 to 17 randomly selected CLL
samples from
the array set of 50, and various normal CD 19+/CD5+ B cells and B and T
lymphocyte
controls by qRT-PCR. An additional set of 3 normal CD19+/CD5+ positive B
cells, not used
for microarray studies, was purchased from AllCells (Berkeley, CA), and used
as an
independent confirmation set. In all instances the qRT-PCR data confirmed the
microarray
data. RNA was treated with RNase-free DNase I and reverse transcribed to cDNA
using
random primers and SuperScript II reverse transcriptase. To determine if the
sense or
21

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
antisense UCR transcript was expressed, total RNA was reverse transcribed
using
Thermoscript RT and a gene specific (i.e. sense or antisense) primer. RT
conditions were as
described (Schmittgen et al., 2004). cDNA was amplified using real-time PCR
and SYBR
green detection using PCR primers designed to amplify the same 40 bp regions
as the oligo
probe on the microarray. The relative amount of each UCR to 18S rRNA was
determined
using the equation 2"dcT, where dCT = (CTUCR- CT18s rRNA) . Relative gene
expression
data were multiplied by 106 to simplify the presentation.
[000142] 4) Northern blot analysis of T-UCRs.
[000143] We analyzed five UCRs, uc.110(N), uc.192(N), uc.246(E), uc.269A(N)
and
uc.352(N), by Northern blot, two of which were then cloned by RACE
experiments. For a
sixth one, the uc. 47(N), the data are not shown. Total RNA was
electrophoresed on 15%
PAA-urea gels (Calin et al., 2002). RNA sources included 11 normal tissues
(breast, liver,
lung, kidney and pancreas) in duplicate or triplicate (purchased from Ambion
and Clontech)
and 4 normal MNC samples and 16 CLL samples prepared in the laboratory. As
this
represents the investigation by Northern blot of UCR expression, we used
multiple samples
from the same tissues to confirm data reproducibility. The probes were
designed to be
identical with the oligonucleotides on the chip in order to detect the same
transcripts as the
microarray, and the hybridization was done as described (Calin et al., 2002).
[000144] B) Databases and Statistical Analyses.
[000145] 1) Databases for Genomic Locations.
[000146] The UCR databases used for all the studies reported here are as
published (Bejerano et
al., 2004b). We restricted our analyses to 481 segments longer than 200 base
pairs (bp). The
Fragile site (FRA) database and the cancer associated genomic regions (CAGR)
databases are
as previously published (Cain et al., 2004b).
[000147] 2) Statistical Analyses for Genomic Locations.
[000148] To test hypotheses associating the incidence of ultra-conserved
regions (UCRs) with
fragile sites, amplified regions in cancer, and deletion regions in cancer, we
utilized random
effect Poisson and negative binomial regression models. Under these models,
"events" were
defined as the number of UCRs, and exposure "time" (i.e. fragile site versus
non-fragile site)
was defined as non-overlapping lengths of the region of interest. The "length"
of a region
was exact, if known, or estimated as 1Mb if unknown. For example, for each
chromosome
22

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
the total length of all non-overlapping fragile sites was computed and was
used as the
exposure time for fragile sites. We then counted the number of UCRs occurring
within
fragile sites for each chromosome. The remaining length of each chromosome
(total Mb -
fragile sites Mb) was assumed to be non-fragile, and the remaining UCRs in
each
chromosome were assumed to occur in the non-fragile region. Then for each
region,
alternative random effects models, the zero-inflated Poisson and the zero-
inflated negative
binomial models were fitted, and, of the three, the best model was selected
using the Akaike's
Information Criteria (based on the log likelihood and number of parameters).
This same
approach was used for analysis of the data from expression of zinc finger
proteins. The best
fitting model for fragile sites with UCRs and LOH with zinc finger proteins
was the zero-
inflated negative binomial. For all other cases, the Poisson model is
reported. When the
number of categories with zero events was more than expected for a Poisson
distribution, the
zero-inflated negative binomial model was preferred. When the total number of
events was
too small for a region, the model likelihoods were unable to converge, and
results are not
reported. The random effect in the Poisson, zero-inflated Poisson and zero-
inflated negative
binomial regression models, was the individual chromosome, in that data within
a
chromosome was assumed to be correlated. The fixed effect in each model
consisted of an
indicator variable(s) for the type of region being compared. We report the
incidence rate
ratio (IRR), 2-sided 95% confidence interval of the incidence rate ratio, and
2-sided p-values
for testing the hypothesis that the incident rate ratio is 1Ø An IRR
significantly > 1 indicates
an increase in the number of UCRs within a region over that expected by
chance.
[000149] The proportions of clustering of miRNAs and zinc finger proteins were
compared
using an asymptotic test of the difference in two independent proportions,
where we report
the difference, 95% confidence interval of the difference, and p-value. Of
note, the ZNF
transcription factor class of genes showed a significantly lower clustering (a
cluster defined
as the location of at least two genes from the same class at less than 50kb
genomic distance)
when compared with the microRNAs (32%, 95/297 clustered ZNF genes versus 48%,
90/186
clustered miRNAs, difference=16.4%, 95% CI=(7.5%, 25.2%), P<0.001).
Allcomputations
were completed using STATA v7.0 and StatXact v7Ø
[000150] 3) Statistical Analyses for negative correlations between microarray
expression
of UCRs and miRNAs.
[000151] A detailed description is provided in the Example II and data
therein. Briefly, the
23

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
input data was constituted by a list of T-UCRs and by a list of miRNAs (the
"seeds") and the
corresponding matrix of expression values. We calculated r, the Spearman rank
coefficient
of correlation for each pair of (miR, UC) genes; namely, we evaluate the P-
values of the
correlation tests and select the genes whose correlation value is significant
at a given value of
rejection. Given the high number of correlation tests performed, P-values were
corrected for
multiple testing by using the false detection rate (FDR), as in (Benjamini and
Hochberg,
1995). In this way, P-values control the number of false positive over the
number of truly
null tests, while FDR controls the number of false positive over the number of
significant
tests.
[000152] C) Functional studies
[000153] 1) UCRs down-regulation by direct interaction with microRNAs.
[000154] The genomic sequences of uc.160, uc.346A and uc.348 were cloned into
pGL3-
control vector (Clontech) using the Xbal site immediately downstream from the
stop codon of
luciferase. Human megakaryocytic MEG-O1 and the cervical carcinoma HeLa cell
lines
were grown as recommended by the ATCC. Cells were co-transfected in 12-well
plates
using siPORT neoFX (Ambion) according to the manufacturer's protocol using 0.4
g of the
firefly luciferase reporter vector and 0.08 g of the control vector
containing Renilla
luciferase, pRL-TK (Promega). For each well 10 nM of miRNA-sense precursor and
scrambled oligonucleotides (Ambion) were used. Firefly and Renilla luciferase
activities
were measured consecutively using the Dual-luciferase assays (Promega) 24 hr
after
transfection. All experiments were performed in triplicate on four to six
different days (n=12
to 18).
[000155] Expression of both the ultraconserved RNA and the mature miRNA was
analyzed
using real-time PCR. Expression of the UCR RNA was determined by real-time PCR
as
described above. Expression of the mature miRNA was performed using TaqMan
looped
primer assays to miR-155 (Applied Biosystems) as described (Chen et al.,
2005). Mature
miRNA expression was presented as 2-dCT where dCT = CTmiRNA - CT1 ss rRNA);
data
was multiplied by 106 to simplify presentation.
[000156] For the patient correlation a set of 13 samples was used (including 9
CLL patients and
4 normal lymphocyte samples) and miR-155, uc.346A and uc.1601evels were
analyzed as
described herein. For the identification of the "in vivo" effects in MEG01 of
miR-155
24

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
transfection, the levels of uc.346A and uc.160 were measured by qRT-PCR as
described at 0,
24 and 48 hrs post-transfection with the pre-miRNA 155 (Ambion) using
Lipofectamine
reagent.
[000157] 2) Effects on cancer cell proliferation by uc. 73A(P) inhibition.
[000158] The siRNA against the uc. 73A(P) were designed using the Dharmacon
algorithm
(Dharmacon siDESIGN (harmacon.com/sidesign)) entering the complete sequence of
the
UCR. The eight highest rank target sequences were tested. The performance was
assessed
after 48, 72 and 144 hour post-transfection by semi-quantitative RT-PCR. The
most effective
two siRNAs and a pool of four different siRNAs, including these two, were
used. We named
these as siRNA1, siRNA3 and siRNApool. For the cell growth assay, the human
colon
cancer cell lines COLO-320 and SW620 were grown in RPMI1640 medium
supplemented
with 10% FBS and 1x104 cells were plated in 96-well plate a day before
transfection. The
cells were transfected with siRNA-uc.73A(P) at a final concentration of 200nM
by using
Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) according to the
manufacturer's
protocol. The siCONTROL Non-Targeting siRNA Pool (Dharmacon Research,
LaFayette,
CO, USA) was used as negative control. The transfection was repeated under the
same
conditions every two days, at 48 and 96 hour. To evaluate the cell number the
CellTiter 96
Aqueous One Solution Cell Proliferation Assay (Promega U.S., Madison, WI, USA)
was
used. The readings were performed at 0, 48, 96, and 144 hours, respectively
measuring the
absorbance at 490nm using an ELISA plate reader (Spectra MAX, Molecular
Devices,
Sunnyvale, CA, USA). The cell growth assay was performed three times in
triplicate for
each treatment. The statistical differences between the number of cells at
various time points
with respect to time 0, was calculated using the t test.
[000159] For both cell cycle and apoptosis assays, cells were plated in 6 well
plates at 6x105
cells per well. The day after and then every 48 hrs, the cells were
transfected with 200nM
siRNA. The cells were collected and fixed in cold 70% ethanol for at least 30
minutes. The
Propidium Iodide (PI) staining was performed at 48, 96, and 144 hours in a 50
g/mL PI
(Sigma Aldrich, St. Louis, MO) and 5.ig/mL RNAse DNAse free (Roche
Diagnostics,
Indianapolis, IN, USA) PBS Solution. The apoptosis staining was performed with
the
Annexin V-FITC Apoptosis Detection Kit (BD Pharmingen, San Jose, CA, USA) and
with
the PE-conjugated monoclonal active Caspase-3 antibody apoptosis kit (BD
Biosciences) at 0
and 144 hours according to the manufacturer's procedure using an FACS Calibur
(BD

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
Biosciences, San Jose, CA, USA) to acquire the data. Each experiment was
performed three
times.
[000160] The GeneBank accession numbers for the cloned T-UCRs described in
this study are
as followings: DQ644536 (UCG.246), DQ644537 (UCG.269A, short form), and
DQ644538
(UCG.269A, long form).
[000161] EXAMPLE II
[000162] Experimental Procedures
[000163] Statistical Analyses for negative correlations between microarray
expression of UCRs
and miRNAs.
[000164] The input data were constituted by a list of UCGs and by a list of
miRNAs (the
"seeds") and the corresponding matrix of expression values. We calculated r,
the Spearman
rank coefficient of correlation, a non-parametric measure of data trend
correlation based on
rankings, for each pair of (miR, UCR) genes; namely, we evaluate the P-values
of the
correlation tests and selected the genes whose correlation value is
significant at a given value
of rejection. Evaluation of P-values was performed assuming that the
correlation values are
distributed using Student's t cumulative distribution, with a number of
degrees of freedom
corresponding to the number of samples in the microarray experiment. The P-
values measure
the 'goodness' of the single correlations (among couples of genes), therefore,
to understand if
the real correlation derives by chance or represents a biologically important
information, we
choose the method of permutations, changing the order of the samples for each
row (miR or
UCR) and calculating the correlations between pair of genes (miR, UCR) with
different
changed samples orders. We repeated the samples permutation and computed
correlations
100 times, in this way, every real correlation has 100 random correlations to
compare with.
Using all (100 * n MIR * n UC) random correlations and real correlations, we
recalculated
P-values based on random correlations ranking and position of the real
correlations.
[000165] Given the high number of correlation tests performed, P-values were
corrected for
multiple testing by using the false detection rate (FDR), as defined by
(Benjamini and
Hochberg, 1995). In this way, P-values control the number of false positive
over the number
of truly null tests, while FDR controls the number of false positive over the
number of
significant tests. Several ways of estimating this number have been proposed,
and we
adopted the solution devised by Tom Nichols (see
froi.sourceforge.net/documents/technicaU
26

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
matlab/FDR), that rescales the P-value obtained on a single test multiplying
it by a
combination of indexes related to the total number of tests performed.
Correction was
performed on a seed by seed basis, meaning that the genes in the seeds list
were considered
independent tests. This statistically validated tripe filtering allows the
targeted extraction of a
shortlist of candidate genes, thus saving resources for the following costly
and time-
consuming genetic analysis.
[000166] To build a scatter plot between miR-24-1 and uc.160 expression
values, we plotted a
regression line by using MatLab function ROBUSTFIT to explain hypotheses of
negative
correlation between these two genes. Notice that only 11.67% (7/60) of points
(pairs of
expression values) are outlier.
[000167] EXAMPLE III
[000168] Additional Examples and Information
[000169] As used herein miR and UCRs are used interchangeably; including in a
non-limiting
manner: a "miR gene product," "microRNA," "miR," or "miRNA" refers to the
unprocessed
(e.g., precursor) or processed (e.g., mature) RNA transcript from a miR gene.
[000170] Diagnosis using UCRs (miRNAs)
[000171] In one aspect, there is provided herein methods of diagnosing whether
a subject has,
or is at risk for developing, a cancer, comprising measuring the level of at
least one UCR in a
test sample from the subject and comparing the level of the miR gene product
in the test
sample to the level of a corresponding miR gene product in a control sample.
As used herein,
a "subject" can be any mammal that has, or is suspected of having, a cancer.
In a preferred
embodiment, the subject is a human who has, or is suspected of having, a
cancer.
[000172] The level of at least one miR gene product can be measured in a
biological sample
(e.g., cells, tissues) obtained from the subject. For example, a tissue sample
(e.g., from a
tumor) can be removed from a subject suspected of having a cancer-related
disease by
conventional biopsy techniques. In another embodiment, a blood sample can be
removed
from the subject, and blood cells (e.g., white blood cells) can be isolated
for DNA extraction
by standard techniques. The blood or tissue sample is preferably obtained from
the subject
prior to initiation of radiotherapy, chemotherapy or other therapeutic
treatment. A
corresponding control tissue or blood sample can be obtained from unaffected
tissues of the
subject, from a normal human individual or population of normal individuals,
or from
27

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
cultured cells corresponding to the majority of cells in the subject's sample.
The control
tissue or blood sample is then processed along with the sample from the
subject, so that the
levels of miR gene product produced from a given miR gene in cells from the
subject's
sample can be compared to the corresponding miR gene product levels from cells
of the
control sample. A reference miR expression standard for the biological sample
can also be
used as a control.
[000173] An alteration (e.g., an increase or decrease) in the level of a miR
gene product in the
sample obtained from the subject, relative to the level of a corresponding miR
gene product
in a control sample, is indicative of the presence of a cancer-related disease
in the subject.
[000174] In one embodiment, the level of the at least one miR gene product in
the test sample is
greater than the level of the corresponding miR gene product in the control
sample (i.e.,
expression of the miR gene product is "up-regulated"). As used herein,
expression of a miR
gene product is "up-regulated" when the amount of miR gene product in a cell
or tissue
sample from a subject is greater than the amount of the same gene product in a
control cell or
tissue sample.
[000175] In another embodiment, the level of the at least one miR gene product
in the test
sample is less than the level of the corresponding miR gene product in the
control sample
(i.e., expression of the miR gene product is "down-regulated"). As used
herein, expression of
a miR gene is "down-regulated" when the amount of miR gene product produced
from that
gene in a cell or tissue sample from a subject is less than the amount
produced from the same
gene in a control cell or tissue sample.
[000176] The relative miR gene expression in the control and normal samples
can be
determined with respect to one or more RNA expression standards. The standards
can
comprise, for example, a zero miR gene expression level, the miR gene
expression level in a
standard cell line, the miR gene expression level in unaffected tissues of the
subject, or the
average level of miR gene expression previously obtained for a population of
normal human
controls.
[000177] The level of a miR gene product in a sample can be measured using any
technique
that is suitable for detecting RNA expression levels in a biological sample.
Suitable
techniques (e.g., Northern blot analysis, RT-PCR, in situ hybridization) for
determining RNA
expression levels in a biological sample (e.g., cells, tissues) are well known
to those of skill
28

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
in the art. In a particular embodiment, the level of at least one miR gene
product is detected
using Northern blot analysis. For example, total cellular RNA can be purified
from cells by
homogenization in the presence of nucleic acid extraction buffer, followed by
centrifugation.
Nucleic acids are precipitated, and DNA is removed by treatment with DNase and
precipitation. The RNA molecules are then separated by gel electrophoresis on
agarose gels
according to standard techniques, and transferred to nitrocellulose filters.
The RNA is then
immobilized on the filters by heating. Detection and quantification of
specific RNA is
accomplished using appropriately labeled DNA or RNA probes complementary to
the RNA
in question. See, for example, Molecular Cloning: A Laboratory Manual, J.
Sambrook et al.,
eds., 2nd edition, Cold Spring Harbor Laboratory Press, 1989, Chapter 7, the
entire disclosure
of which is incorporated by reference.
[000178] Suitable probes for Northern blot hybridization of a given miR gene
product can be
produced from the nucleic acid sequences and include, but are not limited to,
probes having
at least about 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or complete
complementarity to a
miR gene product of interest. Methods for preparation of labeled DNA and RNA
probes, and
the conditions for hybridization thereof to target nucleotide sequences, are
described in
Molecular Cloning: A Laboratory Manual, J. Sambrook et al., eds., 2nd edition,
Cold Spring
Harbor Laboratory Press, 1989, Chapters 10 and 11, the disclosures of which
are incorporated
herein by reference.
[000179] In one non-limiting example, the nucleic acid probe can be labeled
with, e.g., a
radionuclide, such as 3H 32 P 33P 14C, or 35S; a heavy metal; a ligand capable
of functioning
as a specific binding pair member for a labeled ligand (e.g., biotin, avidin
or an antibody); a
fluorescent molecule; a chemiluminescent molecule; an enzyme or the like.
[000180] Probes can be labeled to high specific activity by either the nick
translation method of
Rigby et al. (1977), J. Mol. Biol. 113:237-251 or by the random priming method
of Fienberg
et al. (1983), Anal. Biochem. 132:6-13, the entire disclosures of which are
incorporated
herein by reference. The latter is the method of choice for synthesizing 32P-
labeled probes of
high specific activity from single-stranded DNA or from RNA templates. For
example, by
replacing preexisting nucleotides with highly radioactive nucleotides
according to the nick
translation method, it is possible to prepare 32P-labeled nucleic acid probes
with a specific
activity well in excess of 108 cpm/microgram. Autoradiographic detection of
hybridization
can then be performed by exposing hybridized filters to photographic film.
Densitometric
29

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
scanning of the photographic films exposed by the hybridized filters provides
an accurate
measurement of miR gene transcript levels. Using another approach, miR gene
transcript
levels can be quantified by computerized imaging systems, such as the
Molecular Dynamics
400-B 2D Phosphorima~er available from Amersham Biosciences, Piscataway, NJ.
[000181] Where radionuclide labeling of DNA or RNA probes is not practical,
the random-
primer method can be used to incorporate an analogue, for example, the dTTP
analogue 5-(N-
(N-biotinyl-epsilon-aminocaproyl)-3-aminoallyl) deoxyuridine triphosphate,
into the probe
molecule. The biotinylated probe oligonucleotide can be detected by reaction
with biotin-
binding proteins, such as avidin, streptavidin, and antibodies (e.g., anti-
biotin antibodies)
coupled to fluorescent dyes or enzymes that produce color reactions.
[000182] In addition to Northern and other RNA hybridization techniques,
determining the
levels of RNA transcripts can be accomplished using the technique of in situ
hybridization.
This technique requires fewer cells than the Northern blotting technique, and
involves
depositing whole cells onto a microscope cover slip and probing the nucleic
acid content of
the cell with a solution containing radioactive or otherwise labeled nucleic
acid (e.g., cDNA
or RNA) probes. This technique is particularly well-suited for analyzing
tissue biopsy
samples from subjects. The practice of the in situ hybridization technique is
described in
more detail in U.S. Pat. No. 5,427,916, the entire disclosure of which is
incorporated herein
by reference.
[000183] In one non-limiting example, suitable probes for in situ
hybridization of a given miR
gene product can be produced from the nucleic acid sequences, and include, but
are not
limited to, probes having at least about 70%, 75%, 80%, 85%, 90%, 95%, 98%,
99% or
complete complementarity to a miR gene product of interest, as described
above.
[000184] The relative number of miR gene transcripts in cells can also be
determined by
reverse transcription of miR gene transcripts, followed by amplification of
the reverse-
transcribed transcripts by polymerase chain reaction (RT-PCR). The levels of
miR gene
transcripts can be quantified in comparison with an internal standard, for
example, the level
of mRNA from a "housekeeping" gene present in the same sample. A suitable
"housekeeping" gene for use as an internal standard includes, e.g., myosin or
glyceraldehyde-
3-phosphate dehydrogenase (G3PDH). Methods for performing quantitative and
semi-
quantitative RT-PCR, and variations thereof, are well known to those of skill
in the art.

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000185] In some instances, it may be desirable to simultaneously determine
the expression
level of a plurality of different miR gene products in a sample. In other
instances, it may be
desirable to determine the expression level of the transcripts of all known
miR genes
correlated with a cancer. Assessing cancer-specific expression levels for
hundreds of miR
genes or gene products is time consuming and requires a large amount of total
RNA (e.g., at
least 20 g for each Northern blot) and autoradiographic techniques that
require radioactive
isotopes.
[000186] To overcome these limitations, an oligolibrary, in microchip format
(i.e., a
microarray), may be constructed containing a set of oligonucleotide (e.g.,
oligodeoxynucleotides) probes that are specific for a set of miR genes. Using
such a
microarray, the expression level of multiple microRNAs in a biological sample
can be
determined by reverse transcribing the RNAs to generate a set of target
oligodeoxynucleotides, and hybridizing them to probe the oligonucleotides on
the microarray
to generate a hybridization, or expression, profile. The hybridization profile
of the test
sample can then be compared to that of a control sample to determine which
microRNAs
have an altered expression level in cancer cells.
[000187] As used herein, "probe oligonucleotide" or "probe
oligodeoxynucleotide" refers to an
oligonucleotide that is capable of hybridizing to a target oligonucleotide.
"Target
oligonucleotide" or "target oligodeoxynucleotide" refers to a molecule to be
detected (e.g.,
via hybridization). By "miR-specific probe oligonucleotide" or "probe
oligonucleotide
specific for a miR" is meant a probe oligonucleotide that has a sequence
selected to hybridize
to a specific miR gene product, or to a reverse transcript of the specific miR
gene product.
[000188] An "expression profile" or "hybridization profile" of a particular
sample is essentially
a fingerprint of the state of the sample; while two states may have any
particular gene
similarly expressed, the evaluation of a number of genes simultaneously allows
the
generation of a gene expression profile that is unique to the state of the
cell. That is, normal
tissue may be distinguished from cancerous (e.g., tumor) tissue, and within
cancerous tissue,
different prognosis states (for example, good or poor long term survival
prospects) may be
determined. By comparing expression profiles of the cancer tissue in different
states,
information regarding which genes are important (including both up- and down-
regulation of
genes) in each of these states is obtained. The identification of sequences
that are
differentially expressed in cancer tissue, as well as differential expression
resulting in
31

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
different prognostic outcomes, allows the use of this information in a number
of ways.
[000189] In one non-limiting example, a particular treatment regime may be
evaluated (e.g., to
determine whether a chemotherapeutic drug acts to improve the long-term
prognosis in a
particular patient). Similarly, diagnosis may be done or confirmed by
comparing patient
samples with known expression profiles. Furthermore, these gene expression
profiles (or
individual genes) allow screening of drug candidates that suppress the cancer
expression
profile or convert a poor prognosis profile to a better prognosis profile.
[000190] Accordingly, there is also provided herein methods of diagnosing
whether a subject
has, or is at risk for developing, a cancer, comprising reverse transcribing
RNA from a test
sample obtained from the subject to provide a set of target
oligodeoxynucleotides,
hybridizing the target oligodeoxynucleotides to a microarray comprising miRNA-
specific
probe oligonucleotides to provide a hybridization profile for the test sample,
and comparing
the test sample hybridization profile to a hybridization profile generated
from a control
sample or reference standard, wherein an alteration in the signal of at least
one miRNA is
indicative of the subject either having, or being at risk for developing,
cancer.
[000191] In one embodiment, the microarray comprises miRNA-specific probe
oligonucleotides for a substantial portion of all known human miRNAs. In a
particular
embodiment, the microarray comprises miRNA-specific probe oligonucleotides for
one or
more miRNAs selected from the group consisting of miR29a, miR-29b, miR-29c and
combinations thereof.
[000192] The microarray can be prepared from gene-specific oligonucleotide
probes generated
from known miRNA sequences. The array may contain two different
oligonucleotide probes
for each miRNA, one containing the active, mature sequence and the other being
specific for
the precursor of the miRNA. The array may also contain controls, such as one
or more
mouse sequences differing from human orthologs by only a few bases, which can
serve as
controls for hybridization stringency conditions. tRNAs or other RNAs (e.g.,
rRNAs,
mRNAs) from both species may also be printed on the microchip, providing an
internal,
relatively stable, positive control for specific hybridization. One or more
appropriate controls
for non-specific hybridization may also be included on the microchip. For this
purpose,
sequences are selected based upon the absence of any homology with any known
miRNAs.
[000193] The microarray may be fabricated using techniques known in the art.
For example,
32

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
probe oligonucleotides of an appropriate length, e.g., 40 nucleotides, are 5'-
amine modified
at position C6 and printed using commercially available microarray systems,
e.g., the
GeneMachine OmniGridTm 100 Microarrayer and Amersham CodeLinkTm activated
slides.
Labeled cDNA oligomer corresponding to the target RNAs is prepared by reverse
transcribing the target RNA with labeled primer. Following first strand
synthesis, the
RNA/DNA hybrids are denatured to degrade the RNA templates. The labeled target
cDNAs
thus prepared are then hybridized to the microarray chip under hybridizing
conditions, e.g.,
6X SSPE/30% formamide at 25 C for 18 hours, followed by washing in 0.75X TNT
(Tris
HC1/NaC1/Tween 20) at 37 C for 40 minutes. At positions on the array where the
immobilized probe DNA recognizes a complementary target cDNA in the sample,
hybridization occurs. The labeled target cDNA marks the exact position on the
array where
binding occurs, allowing automatic detection and quantification. The output
consists of a list
of hybridization events, indicating the relative abundance of specific cDNA
sequences, and
therefore the relative abundance of the corresponding complementary miRs, in
the patient
sample.
[000194] According to one embodiment, the labeled cDNA oligomer is a biotin-
labeled cDNA,
prepared from a biotin-labeled primer. The microarray is then processed by
direct detection
of the biotin-containing transcripts using, e.g., Streptavidin-A1exa647
conjugate, and scanned
utilizing conventional scanning methods. Image intensities of each spot on the
array are
proportional to the abundance of the corresponding miR in the patient sample.
[000195] The use of the array has several advantages for miRNA expression
detection. First,
the global expression of several hundred genes can be identified in the same
sample at one
time point. Second, through careful design of the oligonucleotide probes,
expression of both
mature and precursor molecules can be identified. Third, in comparison with
Northern blot
analysis, the chip requires a small amount of RNA, and provides reproducible
results using
2.5 g of total RNA. The relatively limited number of miRNAs (a few hundred
per species)
allows the construction of a common microarray for several species, with
distinct
oligonucleotide probes for each. Such a tool allows for analysis of trans-
species expression
for each known miR under various conditions.
[000196] In addition to use for quantitative expression level assays of
specific miRs, a
microchip containing miRNA-specific probe oligonucleotides corresponding to a
substantial
portion of the miRNome, preferably the entire miRNome, may be employed to
carry out miR
33

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
gene expression profiling, for analysis of miR expression patterns. Distinct
miR signatures
can be associated with established disease markers, or directly with a disease
state.
[000197] According to the expression profiling methods described herein, total
RNA from a
sample from a subject suspected of having a cancer-related disease
quantitatively reverse
transcribed to provide a set of labeled target oligodeoxynucleotides
complementary to the
RNA in the sample. The target oligodeoxynucleotides are then hybridized to a
microarray
comprising miRNA-specific probe oligonucleotides to provide a hybridization
profile for the
sample. The result is a hybridization profile for the sample representing the
expression
pattern of miRNA in the sample. The hybridization profile comprises the signal
from the
binding of the target oligodeoxynucleotides from the sample to the miRNA-
specific probe
oligonucleotides in the microarray. The profile may be recorded as the
presence or absence
of binding (signal vs. zero signal).
[000198] More preferably, the profile recorded includes the intensity of the
signal from each
hybridization. The profile is compared to the hybridization profile generated
from a normal,
i.e., noncancerous, control sample. An alteration in the signal is indicative
of the presence of,
or propensity to develop, cancer in the subject.
[000199] Other techniques for measuring miR gene expression are also within
the skill in the
art, and include various techniques for measuring rates of RNA transcription
and degradation.
[000200] There is also provided herein methods of determining the prognosis of
a subject with
a cancer, comprising measuring the level of at least one miR gene product,
which is
associated with a particular prognosis in a cancer-related disease (e.g., a
good or positive
prognosis, a poor or adverse prognosis), in a test sample from the subject.
[000201] According to these methods, an alteration in the level of a miR gene
product that is
associated with a particular prognosis in the test sample, as compared to the
level of a
corresponding miR gene product in a control sample, is indicative of the
subject having a
cancer with a particular prognosis. In one embodiment, the miR gene product is
associated
with an adverse (i.e., poor) prognosis. Examples of an adverse prognosis
include, but are not
limited to, low survival rate and rapid disease progression. In certain
embodiments, the level
of the at least one miR gene product is measured by reverse transcribing RNA
from a test
sample obtained from the subject to provide a set of target
oligodeoxynucleotides,
hybridizing the target oligodeoxynucleotides to a microarray that comprises
miRNA-specific
34

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
probe oligonucleotides to provide a hybridization profile for the test sample,
and comparing
the test sample hybridization profile to a hybridization profile generated
from a control
sample.
[000202] Without wishing to be bound by any one theory, it is believed that
alterations in the
level of one or more miR gene products in cells can result in the deregulation
of one or more
intended targets for these miRs, which can lead to the formation of cancers.
Therefore,
altering the level of the miR gene product (e.g., by decreasing the level of a
miR gene product
that is up-regulated in cancer cells, by increasing the level of a miR gene
product that is
down-regulated in cancer cells) may successfully treat the cancer.
[000203] Accordingly, there is further provided herein methods of inhibiting
tumorigenesis in a
subject who has, or is suspected of having, a cancer wherein at least one miR
gene product is
deregulated (e.g., down-regulated, up-regulated) in the cancer cells of the
subject. When the
at least one isolated miR gene product is down-regulated in the cancer cells
(e.g., miR-29
family), the method comprises administering an effective amount of the at
least one isolated
miR gene product, or an isolated variant or biologically-active fragment
thereof, such that
proliferation of cancer cells in the subject is inhibited.
[000204] For example, when a miR gene product is down-regulated in a cancer
cell in a subject,
administering an effective amount of an isolated miR gene product to the
subject can inhibit
proliferation of the cancer cell. The isolated miR gene product that is
administered to the
subject can be identical to the endogenous wild-type miR gene product (e.g., a
miR gene
product) that is down-regulated in the cancer cell or it can be a variant or
biologically-active
fragment thereof.
[000205] As defined herein, a "variant" of a miR gene product refers to a
miRNA that has less
than 100% identity to a corresponding wild-type miR gene product and possesses
one or
more biological activities of the corresponding wild-type miR gene product.
Examples of
such biological activities include, but are not limited to, inhibition of
expression of a target
RNA molecule (e.g., inhibiting translation of a target RNA molecule,
modulating the stability
of a target RNA molecule, inhibiting processing of a target RNA molecule) and
inhibition of
a cellular process associated with cancer (e.g., cell differentiation, cell
growth, cell death).
These variants include species variants and variants that are the consequence
of one or more
mutations (e.g., a substitution, a deletion, an insertion) in a miR gene. In
certain
embodiments, the variant is at least about 70%, 75%, 80%, 85%, 90%, 95%, 98%,
or 99%

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
identical to a corresponding wild-type miR gene product.
[000206] As defined herein, a"biologically- active fragment" of a miR gene
product refers to an
RNA fragment of a miR gene product that possesses one or more biological
activities of a
corresponding wild-type miR gene product. As described above, examples of such
biological
activities include, but are not limited to, inhibition of expression of a
target RNA molecule
and inhibition of a cellular process associated with a cancer. In certain
embodiments, the
biologically-active fragment is at least about 5, 7, 10, 12, 15, or 17
nucleotides in length.
[000207] In a particular embodiment, an isolated miR gene product can be
administered to a
subject in combination with one or more additional anti-cancer treatments.
Suitable anti-
cancer treatments include, but are not limited to, chemotherapy, radiation
therapy and
combinations thereof (e.g., chemoradiation).
[000208] When the at least one isolated miR gene product is up-regulated in
the cancer cells,
the method comprises administering to the subject an effective amount of at
least one
compound for inhibiting expression of the at least one miR gene product,
referred to herein as
miR gene expression-inhibition compounds, such that proliferation of the
cancer cells is
inhibited. In a particular embodiment, the at least one miR expression-
inhibition compound
is specific for a miR gene product selected from the group consisting miR29
family,
including miR-29a, miR-29b, miR-29c, and combinations thereof.
[000209] The terms "treat", "treating" and "treatment", as used herein, refer
to ameliorating
symptoms associated with a disease or condition, for example, a cancer,
including preventing
or delaying the onset of the disease symptoms, and/or lessening the severity
or frequency of
symptoms of the disease or condition. The terms "subject", "patient" and
"individual" are
defined herein to include animals, such as mammals, including, but not limited
to, primates,
cows, sheep, goats, horses, dogs, cats, rabbits, guinea pigs, rats, mice or
other bovine, ovine,
equine, canine, feline, rodent, or murine species. In a preferred embodiment,
the animal is a
human.
[000210] As used herein, an "effective amount" of an isolated miR gene product
is an amount
sufficient to inhibit proliferation of a cancer cell in a subject suffering
from a cancer. One
skilled in the art can readily determine an effective amount of a miR gene
product to be
administered to a given subject, by taking into account factors, such as the
size and weight of the
subject; the extent of disease penetration; the age, health and sex of the
subject; the route of
36

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
administration; and whether the administration is regional or systemic.
[000211] For example, an effective amount of an isolated miR gene product can
be based on the
approximate weight of a tumor mass to be treated. The approximate weight of a
tumor mass
can be determined by calculating the approximate volume of the mass, wherein
one cubic
centimeter of volume is roughly equivalent to one gram. An effective amount of
the isolated
miR gene product based on the weight of a tumor mass can be in the range of
about 10-500
micrograms/gram of tumor mass. In certain embodiments, the tumor mass can be
at least
about 10 micrograms/gram of tumor mass, at least about 60 micrograms/gram of
tumor mass
or at least about 100 micrograms/gram of tumor mass.
[000212] An effective amount of an isolated miR gene product can also be based
on the
approximate or estimated body weight of a subject to be treated. Preferably,
such effective
amounts are administered parenterally or enterally, as described herein. For
example, an
effective amount of the isolated miR gene product is administered to a subject
can range from
about 5 to about 3000 micrograms/kg of body weight, from about 700 - 1000
micrograms/kg
of body weight, or greater than about 1000 micrograms/kg of body weight.
[000213] One skilled in the art can also readily determine an appropriate
dosage regimen for the
administration of an isolated miR gene product to a given subject. For
example, a miR gene
product can be administered to the subject once (e.g., as a single injection
or deposition).
Alternatively, a miR gene product can be administered once or twice daily to a
subject for a
period of from about three to about twenty-eight days, more particularly from
about seven to
about ten days. In a particular dosage regimen, a miR gene product is
administered once a
day for seven days. Where a dosage regimen comprises multiple administrations,
it is
understood that the effective amount of the miR gene product administered to
the subject can
comprise the total amount of gene product administered over the entire dosage
regimen.
[000214] As used herein, an "isolated" miR gene product is one that is
synthesized, or altered
or removed from the natural state through human intervention. For example, a
synthetic miR
gene product, or a miR gene product partially or completely separated from the
coexisting
materials of its natural state, is considered to be "isolated." An isolated
miR gene product
can exist in substantially-purified form, or can exist in a cell into which
the miR gene product
has been delivered. Thus, a miR gene product that is deliberately delivered
to, or expressed
in, a cell is considered an "isolated" miR gene product. A miR gene product
produced inside
a cell from a miR precursor molecule is also considered to be an "isolated"
molecule.
37

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
According to one particular embodiment, the isolated miR gene products
described herein
can be used for the manufacture of a medicament for treating a cancer in a
subject (e.g., a
human).
[000215] Isolated miR gene products can be obtained using a number of standard
techniques.
For example, the miR gene products can be chemically synthesized or
recombinantly
produced using methods known in the art. In one embodiment, miR gene products
are
chemically synthesized using appropriately protected ribonucleoside
phosphoramidites and a
conventional DNA/RNA synthesizer. Commercial suppliers of synthetic RNA
molecules or
synthesis reagents include, e.g., Proligo (Hamburg, Germany), Dharmacon
Research
(Lafayette, CO, U.S.A.), Pierce Chemical (part of Perbio Science, Rockford,
IL, U.S.A.),
Glen Research (Sterling, VA, U.S.A.), ChemGenes (Ashland, MA, U.S.A.) and
Cruachem
(Glasgow, UK).
[000216] Alternatively, the miR gene products can be expressed from
recombinant circular or
linear DNA plasmids using any suitable promoter. Suitable promoters for
expressing RNA
from a plasmid include, e.g., the U6 or H1 RNA pol III promoter sequences, or
the
cytomegalovirus promoters. Selection of other suitable promoters is within the
skill in the
art. The recombinant plasmids of the invention can also comprise inducible or
regulatable
promoters for expression of the miR gene products in cancer cells.
[000217] The miR gene products that are expressed from recombinant plasmids
can be isolated
from cultured cell expression systems by standard techniques. The miR gene
products that
are expressed from recombinant plasmids can also be delivered to, and
expressed directly in,
the cancer cells. The use of recombinant plasmids to deliver the miR gene
products to cancer
cells is discussed in more detail below.
[000218] The miR gene products can be expressed from a separate recombinant
plasmid, or
they can be expressed from the same recombinant plasmid. In one embodiment,
the miR
gene products are expressed as RNA precursor molecules from a single plasmid,
and the
precursor molecules are processed into the functional miR gene product by a
suitable
processing system, including, but not limited to, processing systems extant
within a cancer
cell. Other suitable processing systems include, e.g., the in vitro Drosophila
cell lysate
system (e.g., as described in U.S. Published Patent Application No.
2002/0086356 to Tuschl
et al., the entire disclosure of which is incorporated herein by reference)
and the E. coli
RNAse III system (e.g., as described in U.S. Published Patent Application No.
2004/0014113
38

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
to Yang et al., the entire disclosure of which is incorporated herein by
reference).
[000219] Selection of plasmids suitable for expressing the miR gene products,
methods for
inserting nucleic acid sequences into the plasmid to express the gene
products, and methods
of delivering the recombinant plasmid to the cells of interest are within the
skill in the art.
See, for example, Zeng et al. (2002), Molecular Cell 9:1327-1333; Tuschl
(2002), Nat.
Biotechnol, 20:446-448; Brummelkamp et al. (2002), Science 296:550-553;
Miyagishi et al.
(2002), Nat. Biotechnol. 20:497-500; Paddison et al. (2002), Genes Dev. 16:948-
958; Lee et
al. (2002), Nat. Biotechnol. 20:500-505; and Paul et al. (2002), Nat.
Biotechnol. 20:505-508,
the entire disclosures of which are incorporated herein by reference.
[000220] In one embodiment, a plasmid expressing the miR gene products
comprises a
sequence encoding a miR precursor RNA under the control of the CMV
intermediate-early
promoter. As used herein, "under the control" of a promoter means that the
nucleic acid
sequences encoding the miR gene product are located 3' of the promoter, so
that the promoter
can initiate transcription of the miR gene product coding sequences.
[000221] The miR gene products can also be expressed from recombinant viral
vectors. It is
contemplated that the miR gene products can be expressed from two separate
recombinant
viral vectors, or from the same viral vector. The RNA expressed from the
recombinant viral
vectors can either be isolated from cultured cell expression systems by
standard techniques,
or can be expressed directly in cancer cells. The use of recombinant viral
vectors to deliver
the miR gene products to cancer cells is discussed in more detail below.
[000222] The recombinant viral vectors of the invention comprise sequences
encoding the miR
gene products and any suitable promoter for expressing the RNA sequences.
Suitable
promoters include, but are not limited to, the U6 or H1 RNA pol III promoter
sequences, or
the cytomegalovirus promoters. Selection of other suitable promoters is within
the skill in
the art. The recombinant viral vectors of the invention can also comprise
inducible or
regulatable promoters for expression of the miR gene products in a cancer
cell.
[000223] Any viral vector capable of accepting the coding sequences for the
miR gene products
can be used; for example, vectors derived from adenovirus (AV); adeno-
associated virus
(AAV); retroviruses (e.g., lentiviruses (LV), Rhabdoviruses, murine leukemia
virus); herpes
virus, and the like. The tropism of the viral vectors can be modified by
pseudotyping the
vectors with envelope proteins or other surface antigens from other viruses,
or by substituting
39

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
different viral capsid proteins, as appropriate.
[000224] For example, lentiviral vectors of the invention can be pseudotyped
with surface
proteins from vesicular stomatitis virus (VSV), rabies, Ebola, Mokola, and the
like. AAV
vectors of the invention can be made to target different cells by engineering
the vectors to
express different capsid protein serotypes. For example, an AAV vector
expressing a
serotype 2 capsid on a serotype 2 genome is called AAV 2/2. This serotype 2
capsid gene in
the AAV 2/2 vector can be replaced by a serotype 5 capsid gene to produce an
AAV 2/5
vector. Techniques for constructing AAV vectors that express different capsid
protein
serotypes are within the skill in the art; see, e.g., Rabinowitz, J.E., et al.
(2002), J. Virol.
76:791-801, the entire disclosure of which is incorporated herein by
reference.
[000225] Selection of recombinant viral vectors suitable for use in the
invention, methods for
inserting nucleic acid sequences for expressing RNA into the vector, methods
of delivering
the viral vector to the cells of interest, and recovery of the expressed RNA
products are
within the skill in the art. See, for example, Dornburg (1995), Gene Therapy
2:301-310;
Eglitis (1988), Biotechniques 6:608-614; Miller (1990), Hum. Gene Therapy 1:5-
14; and
Anderson (1998), Nature 392:25-30, the entire disclosures of which are
incorporated herein
by reference.
[000226] Particularly suitable viral vectors are those derived from AV and
AAV. A suitable
AV vector for expressing the miR gene products, a method for constructing the
recombinant
AV vector, and a method for delivering the vector into target cells, are
described in Xia et al.
(2002), Nat. Biotech. 20:1006-1010, the entire disclosure of which is
incorporated herein by
reference. Suitable AAV vectors for expressing the miR gene products, methods
for
constructing the recombinant AAV vector, and methods for delivering the
vectors into target
cells are described in Samulski et al. (1987), J. Virol. 61:3096-3101; Fisher
et al. (1996), J.
Virol., 70:520-532; Samulski et al. (1989), J. Virol. 63:3822-3826; U.S. Pat.
No. 5,252,479;
U.S. Pat. No. 5,139,941; International Patent Application No. WO 94/13788; and
International Patent Application No. WO 93/24641, the entire disclosures of
which are
incorporated herein by reference. In one embodiment, the miR gene products are
expressed
from a single recombinant AAV vector comprising the CMV intermediate early
promoter.
[000227] In a certain embodiment, a recombinant AAV viral vector of the
invention comprises
a nucleic acid sequence encoding a miR precursor RNA in operable connection
with a polyT
termination sequence under the control of a human U6 RNA promoter. As used
herein, "in

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
operable connection with a polyT termination sequence" means that the nucleic
acid
sequences encoding the sense or antisense strands are immediately adjacent to
the polyT
termination signal in the 5' direction. During transcription of the miR
sequences from the
vector, the polyT termination signals act to terminate transcription.
[000228] In other embodiments of the treatment methods of the invention, an
effective amount
of at least one compound that inhibits miR expression can be administered to
the subject. As
used herein, "inhibiting miR expression" means that the production of the
precursor and/or
active, mature form of miR gene product after treatment is less than the
amount produced
prior to treatment. One skilled in the art can readily determine whether miR
expression has
been inhibited in a cancer cell, using, for example, the techniques for
determining miR
transcript level discussed above for the diagnostic method. Inhibition can
occur at the level
of gene expression (i.e., by inhibiting transcription of a miR gene encoding
the miR gene
product) or at the level of processing (e.g., by inhibiting processing of a
miR precursor into a
mature, active miR).
[000229] As used herein, an "effective amount" of a compound that inhibits miR
expression is
an amount sufficient to inhibit proliferation of a cancer cell in a subject
suffering from a
cancer (e.g., a cancer). One skilled in the art can readily determine an
effective amount of a
miR expression-inhibition compound to be administered to a given subject, by
taking into
account factors, such as the size and weight of the subject; the extent of
disease penetration; the
age, health and sex of the subject; the route of administration; and whether
the administration is
regional or systemic.
[000230] For example, an effective amount of the expression-inhibition
compound can be based
on the approximate weight of a tumor mass to be treated, as described herein.
An effective
amount of a compound that inhibits miR expression can also be based on the
approximate or
estimated body weight of a subject to be treated, as described herein.
[000231] One skilled in the art can also readily determine an appropriate
dosage regimen for
administering a compound that inhibits miR expression to a given subject.
[000232] Suitable compounds for inhibiting miR gene expression include double-
stranded RNA
(such as short- or small-interfering RNA or "siRNA"), antisense nucleic acids,
and enzymatic
RNA molecules, such as ribozymes. Each of these compounds can be targeted to a
given
miR gene product and interfere with the expression of (e.g., inhibit
translation of, induce
41

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
cleavage or destruction of) the target miR gene product.
[000233] For example, expression of a given miR gene can be inhibited by
inducing RNA
interference of the miR gene with an isolated double-stranded RNA ("dsRNA")
molecule
which has at least 90%, for example at least 95%, at least 98%, at least 99%,
or 100%,
sequence homology with at least a portion of the miR gene product. In a
particular
embodiment, the dsRNA molecule is a "short or small interfering RNA" or
"siRNA."
[000234] siRNA useful in the present methods comprise short double-stranded
RNA from
about 17 nucleotides to about 29 nucleotides in length, preferably from about
19 to about 25
nucleotides in length. The siRNA comprise a sense RNA strand and a
complementary
antisense RNA strand annealed together by standard Watson-Crick base-pairing
interactions
(hereinafter "base-paired"). The sense strand comprises a nucleic acid
sequence that is
substantially identical to a nucleic acid sequence contained within the target
miR gene
product.
[000235] As used herein, a nucleic acid sequence in an siRNA which is
"substantially identical"
to a target sequence contained within the target mRNA is a nucleic acid
sequence that is
identical to the target sequence, or that differs from the target sequence by
one or two
nucleotides. The sense and antisense strands of the siRNA can comprise two
complementary,
single-stranded RNA molecules, or can comprise a single molecule in which two
complementary portions are base-paired and are covalently linked by a single-
stranded
"hairpin" area.
[000236] The siRNA can also be altered RNA that differs from naturally-
occurring RNA by the
addition, deletion, substitution and/or alteration of one or more nucleotides.
Such alterations
can include addition of non-nucleotide material, such as to the end(s) of the
siRNA or to one
or more internal nucleotides of the siRNA, or modifications that make the
siRNA resistant to
nuclease digestion, or the substitution of one or more nucleotides in the
siRNA with
deoxyribonucleotide s .
[000237] One or both strands of the siRNA can also comprise a 3' overhang. As
used herein, a
"3' overhang" refers to at least one unpaired nucleotide extending from the 3'-
end of a
duplexed RNA strand. Thus, in certain embodiments, the siRNA comprises at
least one 3'
overhang of from 1 to about 6 nucleotides (which includes ribonucleotides or
deoxyribonucleotides) in length, from 1 to about 5 nucleotides in length, from
1 to about 4
42

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
nucleotides in length, or from about 2 to about 4 nucleotides in length. In a
particular
embodiment, the 3' overhang is present on both strands of the siRNA, and is 2
nucleotides in
length. For example, each strand of the siRNA can comprise 3' overhangs of
dithymidylic
acid ("TT") or diuridylic acid ("uu").
[000238] The siRNA can be produced chemically or biologically, or can be
expressed from a
recombinant plasmid or viral vector, as described above for the isolated miR
gene products.
Exemplary methods for producing and testing dsRNA or siRNA molecules are
described in
U.S. Published Patent Application No. 2002/0173478 to Gewirtz and in U.S.
Published
Patent Application No. 2004/0018176 to Reich et al., the entire disclosures of
both of which
are incorporated herein by reference.
[000239] Expression of a given miR gene can also be inhibited by an antisense
nucleic acid.
As used herein, an "antisense nucleic acid" refers to a nucleic acid molecule
that binds to
target RNA by means of RNA-RNA, RNA-DNA or RNA-peptide nucleic acid
interactions,
which alters the activity of the target RNA. Antisense nucleic acids suitable
for use in the
present methods are single-stranded nucleic acids (e.g., RNA, DNA, RNA-DNA
chimeras,
peptide nucleic acid (PNA)) that generally comprise a nucleic acid sequence
complementary
to a contiguous nucleic acid sequence in a miR gene product. The antisense
nucleic acid can
comprise a nucleic acid sequence that is 50-100% complementary, 75-100%
complementary,
or 95-100% complementary to a contiguous nucleic acid sequence in a miR gene
product.
[000240] Without wishing to be bound by any theory, it is believed that the
antisense nucleic
acids activate RNase H or another cellular nuclease that digests the miR gene
product/antisense nucleic acid duplex.
[000241] Antisense nucleic acids can also contain modifications to the nucleic
acid backbone or
to the sugar and base moieties (or their equivalent) to enhance target
specificity, nuclease
resistance, delivery or other properties related to efficacy of the molecule.
Such
modifications include cholesterol moieties, duplex intercalators, such as
acridine, or one or
more nuclease-resistant groups.
[000242] Antisense nucleic acids can be produced chemically or biologically,
or can be
expressed from a recombinant plasmid or viral vector, as described above for
the isolated
miR gene products. Exemplary methods for producing and testing are within the
skill in the
art; see, e.g., Stein and Cheng (1993), Science 261:1004 and U.S. Pat. No.
5,849,902 to
43

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
Woolf et al., the entire disclosures of which are incorporated herein by
reference.
[000243] Expression of a given miR gene can also be inhibited by an enzymatic
nucleic acid.
As used herein, an "enzymatic nucleic acid" refers to a nucleic acid
comprising a substrate
binding region that has complementarity to a contiguous nucleic acid sequence
of a miR gene
product, and which is able to specifically cleave the miR gene product. The
enzymatic
nucleic acid substrate binding region can be, for example, 50-100%
complementary, 75-
100% complementary, or 95-100% complementary to a contiguous nucleic acid
sequence in a
miR gene product. The enzymatic nucleic acids can also comprise modifications
at the base,
sugar, and/or phosphate groups.
[000244] Exemplary enzymatic nucleic acids for use in the present methods
include de novo
methyltransferases, including DNMT3A and DNMT3B, as described in the Examples
herein.
[000245] The enzymatic nucleic acids can be produced chemically or
biologically, or can be
expressed from a recombinant plasmid or viral vector, as described above for
the isolated
miR gene products. Exemplary methods for producing and testing dsRNA or siRNA
molecules are described in Werner and Uhlenbeck (1995), Nucl. Acids Res.
23:2092-96;
Hammann et al. (1999), Antisense and Nucleic Acid Drug Dev. 9:25-31; and U.S.
Pat. No.
4,987,071 to Cech et al, the entire disclosures of which are incorporated
herein by reference.
[000246] Administration of at least one miR gene product, or at least one
compound for
inhibiting miR expression, will inhibit the proliferation of cancer cells in a
subject who has a
cancer.
[000247] As used herein, to "inhibit the proliferation of a cancer cell" means
to kill the cell, or
permanently or temporarily arrest or slow the growth of the cell. Inhibition
of cancer cell
proliferation can be inferred if the number of such cells in the subject
remains constant or
decreases after administration of the miR gene products or miR gene expression-
inhibition
compounds. An inhibition of cancer cell proliferation can also be inferred if
the absolute
number of such cells increases, but the rate of tumor growth decreases.
[000248] The number of cancer cells in the body of a subject can be determined
by direct
measurement, or by estimation from the size of primary or metastatic tumor
masses. For
example, the number of cancer cells in a subject can be measured by
immunohistological
methods, flow cytometry, or other techniques designed to detect characteristic
surface
markers of cancer cells.
44

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000249] The size of a tumor mass can be ascertained by direct visual
observation, or by
diagnostic imaging methods, such as X-ray, magnetic resonance imaging,
ultrasound, and
scintigraphy. Diagnostic imaging methods used to ascertain size of the tumor
mass can be
employed with or without contrast agents, as is known in the art. The size of
a tumor mass
can also be ascertained by physical means, such as palpation of the tissue
mass or
measurement of the tissue mass with a measuring instrument, such as a caliper.
[000250] The miR gene products or miR gene expression-inhibition compounds can
be
administered to a subject by any means suitable for delivering these compounds
to cancer
cells of the subject. For example, the miR gene products or miR expression-
inhibition
compounds can be administered by methods suitable to transfect cells of the
subject with
these compounds, or with nucleic acids comprising sequences encoding these
compounds.
[000251] In one embodiment, the cells are transfected with a plasmid or viral
vector comprising
sequences encoding at least one miR gene product or miR gene expression-
inhibition
compound.
[000252] Transfection methods for eukaryotic cells are well known in the art,
and include, e.g.,
direct injection of the nucleic acid into the nucleus or pronucleus of a cell;
electroporation;
liposome transfer or transfer mediated by lipophilic materials; receptor-
mediated nucleic acid
delivery, bioballistic or particle acceleration; calcium phosphate
precipitation, and
transfection mediated by viral vectors.
[000253] For example, cells can be transfected with a liposomal transfer
compound, e.g.,
DOTAP (N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethyl-ammonium methylsulfate,
Boehringer-Mannheim) or an equivalent, such as LIPOFECTIN. The amount of
nucleic acid
used is not critical to the practice of the invention; acceptable results may
be achieved with
0.1-100 micrograms of nucleic acid/105 cells. For example, a ratio of about
0.5 micrograms
of plasmid vector in 3 micrograms of DOTAP per 105 cells can be used.
[000254] A miR gene product or miR gene expression-inhibition compound can
also be
administered to a subject by any suitable enteral or parenteral administration
route. Suitable
enteral administration routes for the present methods include, e.g., oral,
rectal, or intranasal
delivery. Suitable parenteral administration routes include, e.g.,
intravascular administration
(e.g., intravenous bolus injection, intravenous infusion, intra-arterial bolus
injection, intra-
arterial infusion and catheter instillation into the vasculature); peri- and
intra-tissue injection

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
(e.g., peri-tumoral and intra-tumoral injection, intra-retinal injection, or
subretinal injection);
subcutaneous injection or deposition, including subcutaneous infusion (such as
by osmotic
pumps); direct application to the tissue of interest, for example by a
catheter or other
placement device (e.g., a retinal pellet or a suppository or an implant
comprising a porous,
non-porous, or gelatinous material); and inhalation. Particularly suitable
administration
routes are injection, infusion and direct injection into the tumor.
[000255] In the present methods, a miR gene product or miR gene product
expression-
inhibition compound can be administered to the subject either as naked RNA, in
combination
with a delivery reagent, or as a nucleic acid (e.g., a recombinant plasmid or
viral vector)
comprising sequences that express the miR gene product or miR gene product
expression-
inhibition compound. Suitable delivery reagents include, e.g., the Mirus
Transit TKO
lipophilic reagent; lipofectin; lipofectamine; cellfectin; polycations (e.g.,
polylysine), and
liposomes.
[000256] Recombinant plasmids and viral vectors comprising sequences that
express the miR
gene products or miR gene expression-inhibition compounds, and techniques for
delivering
such plasmids and vectors to cancer cells, are discussed herein and/or are
well known in the
art.
[000257] In a particular embodiment, liposomes are used to deliver a miR gene
product or miR
gene expression-inhibition compound (or nucleic acids comprising sequences
encoding them)
to a subject. Liposomes can also increase the blood half-life of the gene
products or nucleic
acids. Suitable liposomes for use in the invention can be formed from standard
vesicle-
forming lipids, which generally include neutral or negatively charged
phospholipids and a
sterol, such as cholesterol. The selection of lipids is generally guided by
consideration of
factors, such as the desired liposome size and half-life of the liposomes in
the blood stream.
A variety of methods are known for preparing liposomes, for example, as
described in Szoka
et al. (1980), Ann. Rev. Biophys. Bioeng. 9:467; and U.S. Pat. Nos. 4,235,871,
4,501,728,
4,837,028, and 5,019,369, the entire disclosures of which are incorporated
herein by
reference.
[000258] The liposomes for use in the present methods can comprise a ligand
molecule that
targets the liposome to cancer cells. Ligands that bind to receptors prevalent
in cancer cells,
such as monoclonal antibodies that bind to tumor cell antigens, are preferred.
46

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000259] The liposomes for use in the present methods can also be modified so
as to avoid
clearance by the mononuclear macrophage system ("MMS") and reticuloendothelial
system
("RES"). Such modified liposomes have opsonization-inhibition moieties on the
surface or
incorporated into the liposome structure. In a particularly preferred
embodiment, a liposome
of the invention can comprise both an opsonization-inhibition moiety and a
ligand.
[000260] Opsonization-inhibiting moieties for use in preparing the liposomes
of the invention
are typically large hydrophilic polymers that are bound to the liposome
membrane. As used
herein, an opsonization-inhibiting moiety is "bound" to a liposome membrane
when it is
chemically or physically attached to the membrane, e.g., by the intercalation
of a lipid-
soluble anchor into the membrane itself, or by binding directly to active
groups of membrane
lipids. These opsonization-inhibiting hydrophilic polymers form a protective
surface layer
that significantly decreases the uptake of the liposomes by the MMS and RES;
e.g., as
described in U.S. Pat. No. 4,920,016, the entire disclosure of which is
incorporated herein by
reference.
[000261] Opsonization-inhibiting moieties suitable for modifying liposomes are
preferably
water-soluble polymers with a number-average molecular weight from about 500
to about
40,000 daltons, and more preferably from about 2,000 to about 20,000 daltons.
Such
polymers include polyethylene glycol (PEG) or polypropylene glycol (PPG)
derivatives; e.g.,
methoxy PEG or PPG, and PEG or PPG stearate; synthetic polymers, such as
polyacrylamide
or poly N-vinyl pyrrolidone; linear, branched, or dendrimeric polyamidoamines;
polyacrylic
acids; polyalcohols, e.g., polyvinylalcohol and polyxylitol to which
carboxylic or amino
groups are chemically linked, as well as gangliosides, such as ganglioside
GM1. Copolymers
of PEG, methoxy PEG, or methoxy PPG, or derivatives thereof, are also
suitable. In
addition, the opsonization-inhibiting polymer can be a block copolymer of PEG
and either a
polyamino acid, polysaccharide, polyamidoamine, polyethyleneamine, or
polynucleotide.
The opsonization-inhibiting polymers can also be natural polysaccharides
containing amino
acids or carboxylic acids, e.g., galacturonic acid, glucuronic acid,
mannuronic acid,
hyaluronic acid, pectic acid, neuraminic acid, alginic acid, carrageenan;
aminated
polysaccharides or oligosaccharides (linear or branched); or carboxylated
polysaccharides or
oligosaccharides, e.g., reacted with derivatives of carbonic acids with
resultant linking of
carboxylic groups. Preferably, the opsonization-inhibiting moiety is a PEG,
PPG, or a
derivative thereof. Liposomes modified with PEG or PEG-derivatives are
sometimes called
47

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
"PEGylated liposomes."
[000262] The opsonization-inhibiting moiety can be bound to the liposome
membrane by any
one of numerous well-known techniques. For example, an N-hydroxysuccinimide
ester of
PEG can be bound to a phosphatidyl-ethanolamine lipid-soluble anchor, and then
bound to a
membrane. Similarly, a dextran polymer can be derivatized with a stearylamine
lipid-soluble
anchor via reductive amination using Na(CN)BH3 and a solvent mixture, such as
tetrahydrofuran and water in a 30:12 ratio at 60 C.
[000263] Liposomes modified with opsonization-inhibition moieties remain in
the circulation
much longer than unmodified liposomes. For this reason, such liposomes are
sometimes
called "stealth" liposomes. Stealth liposomes are known to accumulate in
tissues fed by
porous or "leaky" microvasculature. Thus, tissue characterized by such
microvasculature
defects, for example, tumors, will efficiently accumulate these liposomes; see
Gabizon, et al.
(1988), Proc. Natl. Acad. Sci., U.S.A., 18:6949-53. In addition, the reduced
uptake by the
RES lowers the toxicity of stealth liposomes by preventing significant
accumulation of the
liposomes in the liver and spleen. Thus, liposomes that are modified with
opsonization-
inhibition moieties are particularly suited to deliver the miR gene products
or miR gene
expression-inhibition compounds (or nucleic acids comprising sequences
encoding them) to
tumor cells.
[000264] The miR gene products or miR gene expression-inhibition compounds can
be
formulated as pharmaceutical compositions, sometimes called "medicaments,"
prior to
administering them to a subject, according to techniques known in the art.
Accordingly, the
invention encompasses pharmaceutical compositions for treating a cancer.
[000265] In one embodiment, the pharmaceutical composition comprises at least
one isolated
miR gene product, or an isolated variant or biologically-active fragment
thereof, and a
pharmaceutically-acceptable carrier. In a particular embodiment, the at least
one miR gene
product corresponds to a miR gene product that has a decreased level of
expression in cancer
cells relative to suitable control cells.
[000266] In other embodiments, the pharmaceutical compositions of the
invention comprise at
least one miR expression-inhibition compound. In a particular embodiment, the
at least one
miR gene expression-inhibition compound is specific for a miR gene whose
expression is
greater in cancer cells than control cells.
48

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000267] Pharmaceutical compositions of the present invention are
characterized as being at
least sterile and pyrogen-free. As used herein, "pharmaceutical compositions"
include
formulations for human and veterinary use. Methods for preparing
pharmaceutical
compositions of the invention are within the skill in the art, for example as
described in
Remington's Pharmaceutical Science, 17th ed., Mack Publishing Company, Easton,
Pa.
(1985), the entire disclosure of which is incorporated herein by reference.
[000268] The present pharmaceutical compositions comprise at least one miR
gene product or
miR gene expression-inhibition compound (or at least one nucleic acid
comprising sequences
encoding them) (e.g., 0.1 to 90% by weight), or a physiologically-acceptable
salt thereof,
mixed with a pharmaceutically-acceptable carrier. In certain embodiments, the
pharmaceutical compositions of the invention additionally comprise one or more
anti-cancer
agents (e.g., chemotherapeutic agents). The pharmaceutical formulations of the
invention can
also comprise at least one miR gene product or miR gene expression-inhibition
compound (or
at least one nucleic acid comprising sequences encoding them), which are
encapsulated by
liposomes and a pharmaceutically-acceptable carrier. In one embodiment, the
pharmaceutical
composition comprises a miR gene or gene product that is one or more of
miR29a, miR-29b
and miR-29c.
[000269] Especially suitable pharmaceutically-acceptable carriers are water,
buffered water,
normal saline, 0.4% saline, 0.3% glycine, hyaluronic acid and the like.
[000270] In a particular embodiment, the pharmaceutical compositions of the
invention
comprise at least one miR gene product or miR gene expression-inhibition
compound (or at
least one nucleic acid comprising sequences encoding them) that is resistant
to degradation
by nucleases.
[000271] One skilled in the art can readily synthesize nucleic acids that are
nuclease resistant,
for example, by incorporating one or more ribonucleotides that is modified at
the 2'-position
into the miR gene product. Suitable 2'-modified ribonucleotides include those
modified at
the 2'-position with fluoro, amino, alkyl, alkoxy, and 0-allyl.
[000272] Pharmaceutical Compositions
[000273] Pharmaceutical compositions of the invention can also comprise
conventional
pharmaceutical excipients and/or additives. Suitable pharmaceutical excipients
include
stabilizers, antioxidants, osmolality adjusting agents, buffers, and pH
adjusting agents.
49

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
Suitable additives include, e.g., physiologically biocompatible buffers (e.g.,
tromethamine
hydrochloride), additions of chelants (such as, for example, DTPA or DTPA-
bisamide) or
calcium chelate complexes (such as, for example, calcium DTPA, CaNaDTPA-
bisamide), or,
optionally, additions of calcium or sodium salts (for example, calcium
chloride, calcium
ascorbate, calcium gluconate or calcium lactate). Pharmaceutical compositions
of the
invention can be packaged for use in liquid form, or can be lyophilized.
[000274] For solid pharmaceutical compositions of the invention, conventional
nontoxic solid
pharmaceutically-acceptable carriers can be used; for example, pharmaceutical
grades of
mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum,
cellulose, glucose,
sucrose, magnesium carbonate, and the like.
[000275] For example, a solid pharmaceutical composition for oral
administration can comprise
any of the carriers and excipients listed above and 10-95%, preferably 25%-
75%, of the at
least one miR gene product or miR gene expression-inhibition compound (or at
least one
nucleic acid comprising sequences encoding them). A pharmaceutical composition
for
aerosol (inhalational) administration can comprise 0.01-20% by weight,
preferably 1%-10%
by weight, of the at least one miR gene product or miR gene expression-
inhibition compound
(or at least one nucleic acid comprising sequences encoding them) encapsulated
in a liposome
as described above, and a propellant. A carrier can also be included as
desired; e.g., lecithin
for intranasal delivery.
[000276] The pharmaceutical compositions of the invention can further comprise
one or more
anti-cancer agents. In a particular embodiment, the compositions comprise at
least one miR
gene product or miR gene expression-inhibition compound (or at least one
nucleic acid
comprising sequences encoding them) and at least one chemotherapeutic agent.
Chemotherapeutic agents that are suitable for the methods of the invention
include, but are
not limited to, DNA-alkylating agents, anti-tumor antibiotic agents, anti-
metabolic agents,
tubulin stabilizing agents, tubulin destabilizing agents, hormone antagonist
agents,
topoisomerase inhibitors, protein kinase inhibitors, HMG-CoA inhibitors, CDK
inhibitors,
cyclin inhibitors, caspase inhibitors, metalloproteinase inhibitors, antisense
nucleic acids,
triple-helix DNAs, nucleic acids aptamers, and molecularly-modified viral,
bacterial and
exotoxic agents. Examples of suitable agents for the compositions of the
present invention
include, but are not limited to, cytidine arabinoside, methotrexate,
vincristine, etoposide (VP-
16), doxorubicin (adriamycin), cisplatin (CDDP), dexamethasone, arglabin,

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
cyclophosphamide, sarcolysin, methylnitrosourea, fluorouracil, 5-fluorouracil
(5FU),
vinblastine, camptothecin, actinomycin-D, mitomycin C, hydrogen peroxide,
oxaliplatin,
irinotecan, topotecan, leucovorin, carmustine, streptozocin, CPT- 11, taxol,
tamoxifen,
dacarbazine, rituximab, daunorubicin, 1-(3-D-arabinofuranosylcytosine,
imatinib, fludarabine,
docetaxel, FOLFOX4.
[000277] Inhibitors of Tumorigenesis
[000278] There is also provided herein methods of identifying an inhibitor of
tumorigenesis,
comprising providing a test agent to a cell and measuring the level of at
least one miR gene
product in the cell. In one embodiment, the method comprises providing a test
agent to a cell
and measuring the level of at least one miR gene product associated with
decreased
expression levels in cancer cells. An increase in the level of the miR gene
product in the cell
after the agent is provided, relative to a suitable control cell (e.g., agent
is not provided), is
indicative of the test agent being an inhibitor of tumorigenesis.
[000279] In other embodiments, the method comprises providing a test agent to
a cell and
measuring the level of at least one miR gene product associated with increased
expression
levels in cancer cells. A decrease in the level of the miR gene product in the
cell after the
agent is provided, relative to a suitable control cell (e.g., agent is not
provided), is indicative
of the test agent being an inhibitor of tumorigenesis. In a particular
embodiment, at least one
miR gene product associated with increased expression levels in cancer cells
is selected from
the group consisting of miR29a, miR-29b, miR-29c, and combinations thereof.
[000280] Suitable agents include, but are not limited to drugs (e.g., small
molecules, peptides),
and biological macromolecules (e.g., proteins, nucleic acids). The agent can
be produced
recombinantly, synthetically, or it may be isolated (i.e., purified) from a
natural source.
Various methods for providing such agents to a cell (e.g., transfection) are
well known in the
art, and several of such methods are described hereinabove. Methods for
detecting the
expression of at least one miR gene product (e.g., Northern blotting, in situ
hybridization,
RT-PCR, expression profiling) are also well known in the art. Several of these
methods are
also described hereinabove.
[000281] EXAMPLE IV
[000282] Methods, Reagents and Kits for Diagnosing, Staging, Prognosing,
Monitoring
and Treating Cancer-Related Diseases.
51

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000283] It is to be understood that all examples herein are to be considered
non-limiting in
their scope. Various aspects are described in further detail in the following
subsections.
[000284] Diagnostic Methods
[000285] In one embodiment, there is provided a diagnostic method of assessing
whether a
patient has a cancer-related disease or has higher than normal risk for
developing a cancer-
related disease, comprising the steps of comparing the level of expression of
a marker in a
patient sample and the normal level of expression of the marker in a control,
e.g., a sample
from a patient without a cancer-related disease.
[000286] A significantly higher level of expression of the marker in the
patient sample as
compared to the normal level is an indication that the patient is afflicted
with a cancer-related
disease or has higher than normal risk for developing a cancer-related
disease.
[000287] The markers are selected such that the positive predictive value of
the methods is at
least about 10%, and in certain non-limiting embodiments, about 25%, about 50%
or about
90%. Also preferred for use in the methods are markers that are differentially
expressed, as
compared to normal cells, by at least two-fold in at least about 20%, and in
certain non-
limiting embodiments, about 50% or about 75%.
[000288] In one diagnostic method of assessing whether a patient is afflicted
with a cancer-
related disease (e.g., new detection ("screening"), detection of recurrence,
reflex testing), the
method comprises comparing: a) the level of expression of a marker in a
patient sample, and
b) the normal level of expression of the marker in a control non-cancer-
related disease
sample. A significantly higher level of expression of the marker in the
patient sample as
compared to the normal level is an indication that the patient is afflicted
with a cancer-related
disease.
[000289] There is also provided diagnostic methods for assessing the efficacy
of a therapy for
inhibiting a cancer-related disease in a patient. Such methods comprise
comparing: a)
expression of a marker in a first sample obtained from the patient prior to
providing at least a
portion of the therapy to the patient, and b) expression of the marker in a
second sample
obtained from the patient following provision of the portion of the therapy. A
significantly
lower level of expression of the marker in the second sample relative to that
in the first
sample is an indication that the therapy is efficacious for inhibiting a
cancer-related disease in
the patient.
52

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000290] It will be appreciated that in these methods the "therapy" may be any
therapy for
treating a cancer-related disease including, but not limited to,
pharmaceutical compositions,
gene therapy and biologic therapy such as the administering of antibodies and
chemokines.
Thus, the methods described herein may be used to evaluate a patient before,
during and after
therapy, for example, to evaluate the reduction in disease state.
[000291] In certain aspects, the diagnostic methods are directed to therapy
using a chemical or
biologic agent. These methods comprise comparing: a) expression of a marker in
a first
sample obtained from the patient and maintained in the presence of the
chemical or biologic
agent, and b) expression of the marker in a second sample obtained from the
patient and
maintained in the absence of the agent. A significantly lower level of
expression of the
marker in the second sample relative to that in the first sample is an
indication that the agent
is efficacious for inhibiting a cancer-related disease in the patient. In one
embodiment, the
first and second samples can be portions of a single sample obtained from the
patient or
portions of pooled samples obtained from the patient.
[000292] Methods for Assessing Prognosis
[000293] There is also provided a monitoring method for assessing the
progression of a cancer-
related disease in a patient, the method comprising: a) detecting in a patient
sample at a first
time point, the expression of a marker; b) repeating step a) at a subsequent
time point in time;
and c) comparing the level of expression detected in steps a) and b), and
therefrom
monitoring the progression of a cancer-related disease in the patient. A
significantly higher
level of expression of the marker in the sample at the subsequent time point
from that of the
sample at the first time point is an indication that the cancer-related
disease has progressed,
whereas a significantly lower level of expression is an indication that the
cancer-related
disease has regressed.
[000294] There is further provided a diagnostic method for determining whether
a cancer-
related disease has worsened or is likely to worsen in the future, the method
comprising
comparing: a) the level of expression of a marker in a patient sample, and b)
the normal level
of expression of the marker in a control sample. A significantly higher level
of expression in
the patient sample as compared to the normal level is an indication that the
cancer-related
disease has worsened or is likely to worsen in the future.
[000295] Methods for Assessing Inhibitory, Therapeutic and/or Harmful
Compositions
53

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000296] There is also provided a test method for selecting a composition for
inhibiting a
cancer-related disease in a patient. This method comprises the steps of: a)
obtaining a sample
comprising cells from the patient; b) separately maintaining aliquots of the
sample in the
presence of a plurality of test compositions; c) comparing expression of a
marker in each of
the aliquots; and d) selecting one of the test compositions which
significantly reduces the
level of expression of the marker in the aliquot containing that test
composition, relative to
the levels of expression of the marker in the presence of the other test
compositions.
[000297] There is additionally provided a test method of assessing the harmful
potential of a
compound in causing a cancer-related disease. This method comprises the steps
of: a)
maintaining separate aliquots of cells in the presence and absence of the
compound; and b)
comparing expression of a marker in each of the aliquots. A significantly
higher level of
expression of the marker in the aliquot maintained in the presence of the
compound, relative
to that of the aliquot maintained in the absence of the compound, is an
indication that the
compound possesses such harmful potential.
[000298] In addition, there is further provided a method of inhibiting a
cancer-related disease in
a patient. This method comprises the steps of: a) obtaining a sample
comprising cells from
the patient; b) separately maintaining aliquots of the sample in the presence
of a plurality of
compositions; c) comparing expression of a marker in each of the aliquots; and
d)
administering to the patient at least one of the compositions which
significantly lowers the
level of expression of the marker in the aliquot containing that composition,
relative to the
levels of expression of the marker in the presence of the other compositions.
[000299] The level of expression of a marker in a sample can be assessed, for
example, by
detecting the presence in the sample of: the corresponding marker protein or a
fragment of
the protein (e.g. by using a reagent, such as an antibody, an antibody
derivative, an antibody
fragment or single-chain antibody, which binds specifically with the protein
or protein
fragment) the corresponding marker nucleic acid (e.g. a nucleotide transcript,
or a
complement thereof), or a fragment of the nucleic acid (e.g. by contacting
transcribed
polynucleotides obtained from the sample with a substrate having affixed
thereto one or more
nucleic acids having the entire or a segment of the nucleic acid sequence or a
complement
thereof) a metabolite which is produced directly (i.e., catalyzed) or
indirectly by the
corresponding marker protein.
[000300] Any of the aforementioned methods may be performed using at least one
or a plurality
54

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
(e.g., 2, 3, 5, or 10 or more) of cancer-related disease markers. In such
methods, the level of
expression in the sample of each of a plurality of markers, at least one of
which is a marker,
is compared with the normal level of expression of each of the plurality of
markers in
samples of the same type obtained from control humans not afflicted with a
cancer-related
disease. A significantly altered (i.e., increased or decreased as specified in
the above-
described methods using a single marker) level of expression in the sample of
one or more
markers, or some combination thereof, relative to that marker's corresponding
normal or
control level, is an indication that the patient is afflicted with a cancer-
related disease. For all
of the aforementioned methods, the marker(s) are selected such that the
positive predictive
value of the method is at least about 10%.
[000301] Examples of Candidate Agents
[000302] The candidate agents may be pharmacologic agents already known in the
art or may
be agents previously unknown to have any pharmacological activity. The agents
may be
naturally arising or designed in the laboratory. They may be isolated from
microorganisms,
animals or plants, or may be produced recombinantly, or synthesized by any
suitable
chemical method. They may be small molecules, nucleic acids, proteins,
peptides or
peptidomimetics. In certain embodiments, candidate agents are small organic
compounds
having a molecular weight of more than 50 and less than about 2,500 daltons.
Candidate
agents comprise functional groups necessary for structural interaction with
proteins.
Candidate agents are also found among biomolecules including, but not limited
to: peptides,
saccharides, fatty acids, steroids, purines, pyrimidines, derivatives,
structural analogs or
combinations thereof.
[000303] Candidate agents are obtained from a wide variety of sources
including libraries of
synthetic or natural compounds. There are, for example, numerous means
available for
random and directed synthesis of a wide variety of organic compounds and
biomolecules,
including expression of randomized oligonucleotides and oligopeptides.
Alternatively,
libraries of natural compounds in the form of bacterial, fungal, plant and
animal extracts are
available or readily produced. Additionally, natural or synthetically produced
libraries and
compounds are readily modified through conventional chemical, physical and
biochemical
means, and may be used to produce combinatorial libraries. In certain
embodiments, the
candidate agents can be obtained using any of the numerous approaches in
combinatorial
library methods art, including, by non-limiting example: biological libraries;
spatially

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
addressable parallel solid phase or solution phase libraries; synthetic
library methods
requiring deconvolution; the "one-bead one-compound" library method; and
synthetic library
methods using affinity chromatography selection.
[000304] In certain further embodiments, certain pharmacological agents may be
subjected to
directed or random chemical modifications, such as acylation, alkylation,
esterification,
amidification, etc. to produce structural analogs.
[000305] The same methods for identifying therapeutic agents for treating a
cancer-related
disease can also be used to validate lead compounds/agents generated from in
vitro studies.
[000306] The candidate agent may be an agent that up- or down-regulates one or
more cancer-
related disease response pathways. In certain embodiments, the candidate agent
may be an
antagonist that affects such pathway.
[000307] Methods for Treating a Cancer-related Disease
[000308] There is provided herein methods for treating, inhibiting, relieving
or reversing a
cancer-related disease response. In the methods described herein, an agent
that interferes
with a signaling cascade is administered to an individual in need thereof,
such as, but not
limited to, cancer-related disease patients in whom such complications are not
yet evident and
those who already have at least one cancer-related disease response.
[000309] In the former instance, such treatment is useful to prevent the
occurrence of such
cancer-related disease response and/or reduce the extent to which they occur.
In the latter
instance, such treatment is useful to reduce the extent to which such cancer-
related disease
response occurs, prevent their further development or reverse the cancer-
related disease
response.
[000310] In certain embodiments, the agent that interferes with the cancer-
related disease
response cascade may be an antibody specific for such response.
[000311] Expression of Markers
[000312] Expression of a marker can be inhibited in a number of ways,
including, by way of a
non-limiting example, an antisense oligonucleotide can be provided to the
cancer-related
disease cells in order to inhibit transcription, translation, or both, of the
marker(s).
Alternately, a polynucleotide encoding an antibody, an antibody derivative, or
an antibody
fragment which specifically binds a marker protein, and operably linked with
an appropriate
56

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
promoter/regulator region, can be provided to the cell in order to generate
intracellular
antibodies which will inhibit the function or activity of the protein. The
expression and/or
function of a marker may also be inhibited by treating the cancer-related
disease cell with an
antibody, antibody derivative or antibody fragment that specifically binds a
marker protein.
Using the methods described herein, a variety of molecules, particularly
including molecules
sufficiently small that they are able to cross the cell membrane, can be
screened in order to
identify molecules which inhibit expression of a marker or inhibit the
function of a marker
protein. The compound so identified can be provided to the patient in order to
inhibit cancer-
related disease cells of the patient.
[000313] Any marker or combination of markers, as well as any certain markers
in combination
with the markers, may be used in the compositions, kits and methods described
herein. In
general, it is desirable to use markers for which the difference between the
level of expression
of the marker in cancer-related disease cells and the level of expression of
the same marker in
normal cells is as great as possible. Although this difference can be as small
as the limit of
detection of the method for assessing expression of the marker, it is
desirable that the
difference be at least greater than the standard error of the assessment
method, and, in certain
embodiments, a difference of at least 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, 10-, 15-
, 20-, 100-, 500-,
1000-fold or greater than the level of expression of the same marker in normal
tissue.
[000314] It is recognized that certain marker proteins are secreted to the
extracellular space
surrounding the cells. These markers are used in certain embodiments of the
compositions,
kits and methods, owing to the fact that such marker proteins can be detected
in a cancer-
associated body fluid sample, which may be more easily collected from a human
patient than
a tissue biopsy sample. In addition, in vivo techniques for detection of a
marker protein
include introducing into a subject a labeled antibody directed against the
protein. For
example, the antibody can be labeled with a radioactive marker whose presence
and location
in a subject can be detected by standard imaging techniques.
[000315] In order to determine whether any particular marker protein is a
secreted protein, the
marker protein is expressed in, for example, a mammalian cell, such as a human
cell line,
extracellular fluid is collected, and the presence or absence of the protein
in the extracellular
fluid is assessed (e.g. using a labeled antibody which binds specifically with
the protein).
[000316] It will be appreciated that patient samples containing tissue and/or
fluid cells may be
used in the methods described herein. In these embodiments, the level of
expression of the
57

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
marker can be assessed by assessing the amount (e.g., absolute amount or
concentration) of
the marker in a sample. The cell sample can, of course, be subjected to a
variety of post-
collection preparative and storage techniques (e.g., nucleic acid and/or
protein extraction,
fixation, storage, freezing, ultrafiltration, concentration, evaporation,
centrifugation, etc.)
prior to assessing the amount of the marker in the sample.
[000317] It will also be appreciated that the markers may be shed from the
cells into the
digestive system, the blood stream and/or interstitial spaces. The shed
markers can be tested,
for example, by examining the serum or plasma.
[000318] The compositions, kits and methods can be used to detect expression
of marker
proteins having at least one portion which is displayed on the surface of
cells which express
it. For example, immunological methods may be used to detect such proteins on
whole cells,
or computer-based sequence analysis methods may be used to predict the
presence of at least
one extracellular domain (i.e., including both secreted proteins and proteins
having at least
one cell-surface domain). Expression of a marker protein having at least one
portion which is
displayed on the surface of a cell which expresses it may be detected without
necessarily
lysing the cell (e.g., using a labeled antibody which binds specifically with
a cell-surface
domain of the protein).
[000319] Expression of a marker may be assessed by any of a wide variety of
methods for
detecting expression of a transcribed nucleic acid or protein. Non-limiting
examples of such
methods include immunological methods for detection of secreted, cell-surface,
cytoplasmic
or nuclear proteins, protein purification methods, protein function or
activity assays, nucleic
acid hybridization methods, nucleic acid reverse transcription methods and
nucleic acid
amplification methods.
[000320] In a particular embodiment, expression of a marker is assessed using
an antibody
(e.g., a radio-labeled, chromophore-labeled, fluorophore-labeled or enzyme-
labeled
antibody), an antibody derivative (e.g., an antibody conjugated with a
substrate or with the
protein or ligand of a protein-ligand pair), or an antibody fragment (e.g., a
single-chain
antibody, an isolated antibody hypervariable domain, etc.) which binds
specifically with a
marker protein or fragment thereof, including a marker protein which has
undergone all or a
portion of its normal post-translational modification.
[000321] In another particular embodiment, expression of a marker is assessed
by preparing
58

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
mRNA/cDNA (i.e., a transcribed polynucleotide) from cells in a patient sample,
and by
hybridizing the mRNA/cDNA with a reference polynucleotide which is a
complement of a
marker nucleic acid, or a fragment thereof. cDNA can, optionally, be amplified
using any of
a variety of polymerase chain reaction methods prior to hybridization with the
reference
polynucleotide; preferably, it is not amplified. Expression of one or more
markers can
likewise be detected using quantitative PCR to assess the level of expression
of the marker(s).
Alternatively, any of the many methods of detecting mutations or variants
(e.g., single
nucleotide polymorphisms, deletions, etc.) of a marker may be used to detect
occurrence of a
marker in a patient.
[000322] In a related embodiment, a mixture of transcribed polynucleotides
obtained from the
sample is contacted with a substrate having fixed thereto a polynucleotide
complementary to
or homologous with at least a portion (e.g., at least 7, 10, 15, 20, 25, 30,
40, 50, 100, 500, or
more nucleotide residues) of a marker nucleic acid. If polynucleotides
complementary to or
homologous with are differentially detectable on the substrate (e.g.,
detectable using different
chromophores or fluorophores, or fixed to different selected positions), then
the levels of
expression of a plurality of markers can be assessed simultaneously using a
single substrate
(e.g., a "gene chip" microarray of polynucleotides fixed at selected
positions). When a
method of assessing marker expression is used which involves hybridization of
one nucleic
acid with another, it is desired that the hybridization be performed under
stringent
hybridization conditions.
[000323] Biomarker Assays
[000324] In certain embodiments, the biomarker assays can be performed using
mass
spectrometry or surface plasmon resonance. In various embodiment, the method
of
identifying an agent active against a 1 cancer-related disease can include a)
providing a
sample of cells containing one or more markers or derivative thereof; b)
preparing an extract
from said cells; c) mixing said extract with a labeled nucleic acid probe
containing a marker
binding site; and, d) determining the formation of a complex between the
marker and the
nucleic acid probe in the presence or absence of the test agent. The
determining step can
include subjecting said extract/nucleic acid probe mixture to an
electrophoretic mobility shift
assay.
[000325] In certain embodiments, the determining step comprises an assay
selected from an
enzyme linked immunoabsorption assay (ELISA), fluorescence based assays and
ultra high
59

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
throughput assays, for example surface plasmon resonance (SPR) or fluorescence
correlation
spectroscopy (FCS) assays. In such embodiments, the SPR sensor is useful for
direct real-
time observation of biomolecular interactions since SPR is sensitive to minute
refractive
index changes at a metal-dielectric surface. SPR is a surface technique that
is sensitive to
changes of 105 to 10-6 refractive index (RI) units within approximately 200 nm
of the SPR
sensor/sample interface. Thus, SPR spectroscopy is useful for monitoring the
growth of thin
organic films deposited on the sensing layer.
[000326] Because the compositions, kits, and methods rely on detection of a
difference in
expression levels of one or more markers, it is desired that the level of
expression of the
marker is significantly greater than the minimum detection limit of the method
used to assess
expression in at least one of normal cells and cancer-affected cells.
[000327] It is understood that by routine screening of additional patient
samples using one or
more of the markers, it will be realized that certain of the markers are over-
expressed in cells
of various types, including specific cancer-related diseases.
[000328] In addition, as a greater number of patient samples are assessed for
expression of the
markers and the outcomes of the individual patients from whom the samples were
obtained
are correlated, it will also be confirmed that altered expression of certain
of the markers are
strongly correlated with a cancer-related disease and that altered expression
of other markers
are strongly correlated with other diseases. The compositions, kits, and
methods are thus
useful for characterizing one or more of the stage, grade, histological type,
and nature of a
cancer-related disease in patients.
[000329] When the compositions, kits, and methods are used for characterizing
one or more of
the stage, grade, histological type, and nature of a cancer-related disease in
a patient, it is
desired that the marker or panel of markers is selected such that a positive
result is obtained
in at least about 20%, and in certain embodiments, at least about 40%, 60%, or
80%, and in
substantially all patients afflicted with a cancer-related disease of the
corresponding stage,
grade, histological type, or nature. The marker or panel of markers invention
can be selected
such that a positive predictive value of greater than about 10% is obtained
for the general
population (in a non-limiting example, coupled with an assay specificity
greater than 80%).
[000330] When a plurality of markers are used in the compositions, kits, and
methods, the level
of expression of each marker in a patient sample can be compared with the
normal level of

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
expression of each of the plurality of markers in non-cancer samples of the
same type, either
in a single reaction mixture (i.e. using reagents, such as different
fluorescent probes, for each
marker) or in individual reaction mixtures corresponding to one or more of the
markers. In
one embodiment, a significantly increased level of expression of more than one
of the
plurality of markers in the sample, relative to the corresponding normal
levels, is an
indication that the patient is afflicted with a cancer-related disease. When a
plurality of
markers is used, 2, 3, 4, 5, 8, 10, 12, 15, 20, 30, or 50 or more individual
markers can be
used; in certain embodiments, the use of fewer markers may be desired.
[000331] In order to maximize the sensitivity of the compositions, kits, and
methods (i.e. by
interference attributable to cells of non-tissue and/or fluid origin in a
patient sample), it is
desirable that the marker used therein be a marker which has a restricted
tissue distribution,
e.g., normally not expressed in a non-tissue cells.
[000332] It is recognized that the compositions, kits, and methods will be of
particular utility to
patients having an enhanced risk of developing a cancer-related disease and
their medical
advisors. Patients recognized as having an enhanced risk of developing a
cancer-related
disease include, for example, patients having a familial history of a cancer-
related disease.
[000333] The level of expression of a marker in normal human cells can be
assessed in a variety
of ways. In one embodiment, this normal level of expression is assessed by
assessing the
level of expression of the marker in a portion of cells which appear to be
normal and by
comparing this normal level of expression with the level of expression in a
portion of the
cells which is suspected of being abnormal. Alternately, and particularly as
further
information becomes available as a result of routine performance of the
methods described
herein, population-average values for normal expression of the markers may be
used. In
other embodiments, the "normal" level of expression of a marker may be
determined by
assessing expression of the marker in a patient sample obtained from a non-
cancer-afflicted
patient, from a patient sample obtained from a patient before the suspected
onset of a cancer-
related disease in the patient, from archived patient samples, and the like.
[000334] There is also provided herein compositions, kits, and methods for
assessing the
presence of cancer-related disease cells in a sample (e.g. an archived tissue
sample or a
sample obtained from a patient). These compositions, kits, and methods are
substantially the
same as those described above, except that, where necessary, the compositions,
kits, and
methods are adapted for use with samples other than patient samples. For
example, when the
61

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
sample to be used is a parafinized, archived human tissue sample, it can be
necessary to
adjust the ratio of compounds in the compositions, in the kits, or the methods
used to assess
levels of marker expression in the sample.
[000335] Methods of Producing Antibodies
[000336] There is also provided herein a method of making an isolated
hybridoma which
produces an antibody useful for assessing whether a patient is afflicted with
a cancer-related
disease. In this method, a protein or peptide comprising the entirety or a
segment of a marker
protein is synthesized or isolated (e.g. by purification from a cell in which
it is expressed or
by transcription and translation of a nucleic acid encoding the protein or
peptide in vivo or in
vitro). A vertebrate, for example, a mammal such as a mouse, rat, rabbit, or
sheep, is
immunized using the protein or peptide. The vertebrate may optionally (and
preferably) be
immunized at least one additional time with the protein or peptide, so that
the vertebrate
exhibits a robust immune response to the protein or peptide. Splenocytes are
isolated from
the immunized vertebrate and fused with an immortalized cell line to form
hybridomas, using
any of a variety of methods. Hybridomas formed in this manner are then
screened using
standard methods to identify one or more hybridomas which produce an antibody
which
specifically binds with the marker protein or a fragment thereof. There is
also provided
herein hybridomas made by this method and antibodies made using such
hybridomas.
[000337] Methods of Assessing Efficacy
[000338] There is also provided herein a method of assessing the efficacy of a
test compound
for inhibiting cancer-related disease cells. As described herein, differences
in the level of
expression of the markers correlate with the abnormal state of the cells.
Although it is
recognized that changes in the levels of expression of certain of the markers
likely result from
the abnormal state of the cells, it is likewise recognized that changes in the
levels of
expression of other of the markers induce, maintain, and promote the abnormal
state of those
cells. Thus, compounds which inhibit a cancer-related disease in a patient
will cause the
level of expression of one or more of the markers to change to a level nearer
the normal level
of expression for that marker (i.e. the level of expression for the marker in
normal cells).
[000339] This method thus comprises comparing expression of a marker in a
first cell sample
and maintained in the presence of the test compound and expression of the
marker in a
second cell sample and maintained in the absence of the test compound. A
significantly
62

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
reduced expression of a marker in the presence of the test compound is an
indication that the
test compound inhibits a cancer-related disease. The cell samples may, for
example, be
aliquots of a single sample of normal cells obtained from a patient, pooled
samples of normal
cells obtained from a patient, cells of a normal cell line, aliquots of a
single sample of cancer-
related disease cells obtained from a patient, pooled samples of cancer-
related disease cells
obtained from a patient, cells of a cancer-related disease cell line, or the
like.
[000340] In one embodiment, the samples are cancer-related disease cells
obtained from a
patient and a plurality of compounds believed to be effective for inhibiting
various cancer-
related diseases are tested in order to identify the compound which is likely
to best inhibit the
cancer-related disease in the patient.
[000341] This method may likewise be used to assess the efficacy of a therapy
for inhibiting a
cancer-related disease in a patient. In this method, the level of expression
of one or more
markers in a pair of samples (one subjected to the therapy, the other not
subjected to the
therapy) is assessed. As with the method of assessing the efficacy of test
compounds, if the
therapy induces a significantly lower level of expression of a marker then the
therapy is
efficacious for inhibiting a cancer-related disease. As above, if samples from
a selected
patient are used in this method, then alternative therapies can be assessed in
vitro in order to
select a therapy most likely to be efficacious for inhibiting a cancer-related
disease in the
patient.
[000342] Methods for Assessing Harmful Potentials
[000343] As described herein, the abnormal state of human cells is correlated
with changes in
the levels of expression of the markers. There is also provided a method for
assessing the
harmful potential of a test compound. This method comprises maintaining
separate aliquots
of human cells in the presence and absence of the test compound. Expression of
a marker in
each of the aliquots is compared. A significantly higher level of expression
of a marker in the
aliquot maintained in the presence of the test compound (relative to the
aliquot maintained in
the absence of the test compound) is an indication that the test compound
possesses a harmful
potential. The relative harmful potential of various test compounds can be
assessed by
comparing the degree of enhancement or inhibition of the level of expression
of the relevant
markers, by comparing the number of markers for which the level of expression
is enhanced
or inhibited, or by comparing both.
63

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000344] Isolated Proteins and Antibodies
[000345] One aspect pertains to isolated marker proteins and biologically
active portions
thereof, as well as polypeptide fragments suitable for use as immunogens to
raise antibodies
directed against a marker protein or a fragment thereof. In one embodiment,
the native
marker protein can be isolated from cells or tissue sources by an appropriate
purification
scheme using standard protein purification techniques. In another embodiment,
a protein or
peptide comprising the whole or a segment of the marker protein is produced by
recombinant
DNA techniques. Alternative to recombinant expression, such protein or peptide
can be
synthesized chemically using standard peptide synthesis techniques.
[000346] An "isolated" or "purified" protein or biologically active portion
thereof is
substantially free of cellular material or other contaminating proteins from
the cell or tissue
source from which the protein is derived, or substantially free of chemical
precursors or other
chemicals when chemically synthesized. The language "substantially free of
cellular
material" includes preparations of protein in which the protein is separated
from cellular
components of the cells from which it is isolated or recombinantly produced.
Thus, protein
that is substantially free of cellular material includes preparations of
protein having less than
about 30%, 20%, 10%, or 5% (by dry weight) of heterologous protein (also
referred to herein
as a "contaminating protein").
[000347] When the protein or biologically active portion thereof is
recombinantly produced, it
is also preferably substantially free of culture medium, i.e., culture medium
represents less
than about 20%, 10%, or 5% of the volume of the protein preparation. When the
protein is
produced by chemical synthesis, it is preferably substantially free of
chemical precursors or
other chemicals, i.e., it is separated from chemical precursors or other
chemicals which are
involved in the synthesis of the protein. Accordingly such preparations of the
protein have
less than about 30%, 20%, 10%, 5% (by dry weight) of chemical precursors or
compounds
other than the polypeptide of interest.
[000348] Biologically active portions of a marker protein include polypeptides
comprising
amino acid sequences sufficiently identical to or derived from the amino acid
sequence of the
marker protein, which include fewer amino acids than the full length protein,
and exhibit at
least one activity of the corresponding full-length protein. Typically,
biologically active
portions comprise a domain or motif with at least one activity of the
corresponding full-
length protein. A biologically active portion of a marker protein can be a
polypeptide which
64

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
is, for example, 10, 25, 50, 100 or more amino acids in length. Moreover,
other biologically
active portions, in which other regions of the marker protein are deleted, can
be prepared by
recombinant techniques and evaluated for one or more of the functional
activities of the
native form of the marker protein. In certain embodiments, useful proteins are
substantially
identical (e.g., at least about 40%, and in certain embodiments, 50%, 60%,
70%, 80%, 90%,
95%, or 99%) to one of these sequences and retain the functional activity of
the
corresponding naturally-occurring marker protein yet differ in amino acid
sequence due to
natural allelic variation or mutagenesis.
[000349] In addition, libraries of segments of a marker protein can be used to
generate a
variegated population of polypeptides for screening and subsequent selection
of variant
marker proteins or segments thereof.
[000350] Predictive Medicine
[000351] There is also provided herein uses of the animal models and markers
in the field of
predictive medicine in which diagnostic assays, prognostic assays,
pharmacogenomics, and
monitoring clinical trials are used for prognostic (predictive) purposes to
thereby treat an
individual prophylactically. Accordingly, there is also provided herein
diagnostic assays for
determining the level of expression of one or more marker proteins or nucleic
acids, in order
to determine whether an individual is at risk of developing a cancer-related
disease. Such
assays can be used for prognostic or predictive purposes to thereby
prophylactically treat an
individual prior to the onset of the cancer-related disease.
[000352] In another aspect, the methods are useful for at least periodic
screening of the same
individual to see if that individual has been exposed to chemicals or toxins
that change
his/her expression patterns.
[000353] Yet another aspect pertains to monitoring the influence of agents
(e.g., drugs or other
compounds administered either to inhibit a cancer-related disease or to treat
or prevent any
other disorder (e.g., in order to understand any system effects that such
treatment may have)
on the expression or activity of a marker in clinical trials.
[000354] Pharmacogenomics
[000355] The markers are also useful as pharmacogenomic markers. As used
herein, a
"pharmacogenomic marker" is an objective biochemical marker whose expression
level
correlates with a specific clinical drug response or susceptibility in a
patient. The presence or

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
quantity of the pharmacogenomic marker expression is related to the predicted
response of
the patient and more particularly the patient's tumor to therapy with a
specific drug or class of
drugs. By assessing the presence or quantity of the expression of one or more
pharmacogenomic markers in a patient, a drug therapy which is most appropriate
for the
patient, or which is predicted to have a greater degree of success, may be
selected.
[000356] Monitoring Clinical Trials
[000357] Monitoring the influence of agents (e.g., drug compounds) on the
level of expression
of a marker can be applied not only in basic drug screening, but also in
clinical trials. For
example, the effectiveness of an agent to affect marker expression can be
monitored in
clinical trials of subjects receiving treatment for a cancer-related disease.
[000358] In one non-limiting embodiment, the present invention provides a
method for
monitoring the effectiveness of treatment of a subject with an agent (e.g., an
agonist,
antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or
other drug
candidate) comprising the steps of (i) obtaining a pre-administration sample
from a subject
prior to administration of the agent; (ii) detecting the level of expression
of one or more
selected markers in the pre-administration sample; (iii) obtaining one or more
post-
administration samples from the subject; (iv) detecting the level of
expression of the
marker(s) in the post-administration samples; (v) comparing the level of
expression of the
marker(s) in the pre-administration sample with the level of expression of the
marker(s) in the
post-administration sample or samples; and (vi) altering the administration of
the agent to the
subject accordingly.
[000359] For example, increased expression of the marker gene(s) during the
course of
treatment may indicate ineffective dosage and the desirability of increasing
the dosage.
Conversely, decreased expression of the marker gene(s) may indicate
efficacious treatment
and no need to change dosage.
[000360] Electronic Apparatus Readable Media, Systems, Arrays and Methods of
Using
Same
[000361] As used herein, "electronic apparatus readable media" refers to any
suitable medium
for storing, holding or containing data or information that can be read and
accessed directly
by an electronic apparatus. Such media can include, but are not limited to:
magnetic storage
media, such as floppy discs, hard disc storage medium, and magnetic tape;
optical storage
66

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
media such as compact disc; electronic storage media such as RAM, ROM, EPROM,
EEPROM and the like; and general hard disks and hybrids of these categories
such as
magnetic/optical storage media. The medium is adapted or configured for having
recorded
thereon a marker as described herein.
[000362] As used herein, the term "electronic apparatus" is intended to
include any suitable
computing or processing apparatus or other device configured or adapted for
storing data or
information. Examples of electronic apparatus suitable for use with the
present invention
include stand-alone computing apparatus; networks, including a local area
network (LAN), a
wide area network (WAN) Internet, Intranet, and Extranet; electronic
appliances such as
personal digital assistants (PDAs), cellular phone, pager and the like; and
local and
distributed processing systems.
[000363] As used herein, "recorded" refers to a process for storing or
encoding information on
the electronic apparatus readable medium. Those skilled in the art can readily
adopt any
method for recording information on media to generate materials comprising the
markers
described herein.
[000364] A variety of software programs and formats can be used to store the
marker
information of the present invention on the electronic apparatus readable
medium. Any
number of data processor structuring formats (e.g., text file or database) may
be employed in
order to obtain or create a medium having recorded thereon the markers. By
providing the
markers in readable form, one can routinely access the marker sequence
information for a
variety of purposes. For example, one skilled in the art can use the
nucleotide or amino acid
sequences in readable form to compare a target sequence or target structural
motif with the
sequence information stored within the data storage means. Search means are
used to
identify fragments or regions of the sequences which match a particular target
sequence or
target motif.
[000365] Thus, there is also provided herein a medium for holding instructions
for performing a
method for determining whether a subject has a cancer-related disease or a pre-
disposition to
a cancer-related disease, wherein the method comprises the steps of
determining the presence
or absence of a marker and based on the presence or absence of the marker,
determining
whether the subject has a cancer-related disease or a pre-disposition to a
cancer-related
disease and/or recommending a particular treatment for a cancer-related
disease or pre-
cancer-related disease condition.
67

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000366] There is also provided herein an electronic system and/or in a
network, a method for
determining whether a subject has a cancer-related disease or a pre-
disposition to a cancer-
related disease associated with a marker wherein the method comprises the
steps of
determining the presence or absence of the marker, and based on the presence
or absence of
the marker, determining whether the subject has a cancer-related disease or a
pre-disposition
to a cancer-related disease, and/or recommending a particular treatment for
the cancer-related
disease or pre-cancer-related disease condition. The method may further
comprise the step of
receiving phenotypic information associated with the subject and/or acquiring
from a network
phenotypic information associated with the subject.
[000367] Also provided herein is a network, a method for determining whether a
subject has a
cancer-related disease or a pre-disposition to a cancer-related disease
associated with a
marker, the method comprising the steps of receiving information associated
with the marker,
receiving phenotypic information associated with the subject, acquiring
information from the
network corresponding to the marker and/or a cancer-related disease, and based
on one or
more of the phenotypic information, the marker, and the acquired information,
determining
whether the subject has a cancer-related disease or a pre-disposition to a
cancer-related
disease. The method may further comprise the step of recommending a particular
treatment
for the cancer-related disease or pre-cancer-related disease condition.
[000368] There is also provided herein a business method for determining
whether a subject has
a cancer-related disease or a pre-disposition to a cancer-related disease, the
method
comprising the steps of receiving information associated with the marker,
receiving
phenotypic information associated with the subject, acquiring information from
the network
corresponding to the marker and/or a cancer-related disease, and based on one
or more of the
phenotypic information, the marker, and the acquired information, determining
whether the
subject has a cancer-related disease or a pre-disposition to a cancer-related
disease. The
method may further comprise the step of recommending a particular treatment
for the cancer-
related disease or pre-cancer-related disease condition.
[000369] Arrays
[000370] There is also provided herein an array that can be used to assay
expression of one or
more genes in the array. In one embodiment, the array can be used to assay
gene expression
in a tissue to ascertain tissue specificity of genes in the array. In this
manner, up to about
7000 or more genes can be simultaneously assayed for expression. This allows a
profile to be
68

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
developed showing a battery of genes specifically expressed in one or more
tissues.
[000371] In addition to such qualitative determination, there is provided
herein the quantitation
of gene expression. Thus, not only tissue specificity, but also the level of
expression of a
battery of genes in the tissue is ascertainable. Thus, genes can be grouped on
the basis of
their tissue expression per se and level of expression in that tissue. This is
useful, for
example, in ascertaining the relationship of gene expression between or among
tissues. Thus,
one tissue can be perturbed and the effect on gene expression in a second
tissue can be
determined. In this context, the effect of one cell type on another cell type
in response to a
biological stimulus can be determined.
[000372] Such a determination is useful, for example, to know the effect of
cell-cell interaction
at the level of gene expression. If an agent is administered therapeutically
to treat one cell
type but has an undesirable effect on another cell type, the method provides
an assay to
determine the molecular basis of the undesirable effect and thus provides the
opportunity to
co-administer a counteracting agent or otherwise treat the undesired effect.
Similarly, even
within a single cell type, undesirable biological effects can be determined at
the molecular
level. Thus, the effects of an agent on expression of other than the target
gene can be
ascertained and counteracted.
[000373] In another embodiment, the array can be used to monitor the time
course of
expression of one or more genes in the array. This can occur in various
biological contexts,
as disclosed herein, for example development of a cancer-related disease,
progression of a
cancer-related disease, and processes, such as cellular transformation
associated with a
cancer-related disease.
[000374] The array is also useful for ascertaining the effect of the
expression of a gene or the
expression of other genes in the same cell or in different cells. This
provides, for example,
for a selection of alternate molecular targets for therapeutic intervention if
the ultimate or
downstream target cannot be regulated.
[000375] The array is also useful for ascertaining differential expression
patterns of one or
more genes in normal and abnormal cells. This provides a battery of genes that
could serve
as a molecular target for diagnosis or therapeutic intervention.
[000376] Surrogate Markers
[000377] The markers may serve as surrogate markers for one or more disorders
or disease
69

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
states or for conditions leading up to a cancer-related disease state. As used
herein, a
"surrogate marker" is an objective biochemical marker which correlates with
the absence or
presence of a disease or disorder, or with the progression of a disease or
disorder. The
presence or quantity of such markers is independent of the disease. Therefore,
these markers
may serve to indicate whether a particular course of treatment is effective in
lessening a
disease state or disorder. Surrogate markers are of particular use when the
presence or extent
of a disease state or disorder is difficult to assess through standard
methodologies, or when an
assessment of disease progression is desired before a potentially dangerous
clinical endpoint
is reached.
[000378] The markers are also useful as pharmacodynamic markers. As used
herein, a
"pharmacodynamic marker" is an objective biochemical marker which correlates
specifically
with drug effects. The presence or quantity of a pharmacodynamic marker is not
related to
the disease state or disorder for which the drug is being administered;
therefore, the presence
or quantity of the marker is indicative of the presence or activity of the
drug in a subject. For
example, a pharmacodynamic marker may be indicative of the concentration of
the drug in a
biological tissue, in that the marker is either expressed or transcribed or
not expressed or
transcribed in that tissue in relationship to the level of the drug. In this
fashion, the
distribution or uptake of the drug may be monitored by the pharmacodynamic
marker.
Similarly, the presence or quantity of the pharmacodynamic marker may be
related to the
presence or quantity of the metabolic product of a drug, such that the
presence or quantity of
the marker is indicative of the relative breakdown rate of the drug in vivo.
[000379] Pharmacodynamic markers are of particular use in increasing the
sensitivity of
detection of drug effects, particularly when the drug is administered in low
doses. Since even
a small amount of a drug may be sufficient to activate multiple rounds of
marker transcription
or expression, the amplified marker may be in a quantity which is more readily
detectable
than the drug itself. Also, the marker may be more easily detected due to the
nature of the
marker itself; for example, using the methods described herein, antibodies may
be employed
in an immune-based detection system for a protein marker, or marker-specific
radiolabeled
probes may be used to detect a mRNA marker. Furthermore, the use of a
pharmacodynamic
marker may offer mechanism-based prediction of risk due to drug treatment
beyond the range
of possible direct observations.
[000380] Protocols for Testing

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000381] The method of testing for cancer-related diseases comprises, for
example measuring
the expression level of each marker gene in a biological sample from a subject
over time and
comparing the level with that of the marker gene in a control biological
sample.
[000382] When the marker gene is one of the genes described herein and the
expression level is
differentially expressed (for examples, higher or lower than that in the
control), the subject is
judged to be affected with a cancer-related disease. When the expression level
of the marker
gene falls within the permissible range, the subject is unlikely to be
affected with a cancer-
related disease.
[000383] The standard value for the control may be pre-determined by measuring
the
expression level of the marker gene in the control, in order to compare the
expression levels.
For example, the standard value can be determined based on the expression
level of the
above-mentioned marker gene in the control. For example, in certain
embodiments, the
permissible range is taken as 2S.D. based on the standard value. Once the
standard value is
determined, the testing method may be performed by measuring only the
expression level in a
biological sample from a subject and comparing the value with the determined
standard value
for the control.
[000384] Expression levels of marker genes include transcription of the marker
genes to
mRNA, and translation into proteins. Therefore, one method of testing for a
cancer-related
disease is performed based on a comparison of the intensity of expression of
mRNA
corresponding to the marker genes, or the expression level of proteins encoded
by the marker
genes.
[000385] Probes
[000386] The measurement of the expression levels of marker genes in the
testing for a cancer-
related disease can be carried out according to various gene analysis methods.
Specifically,
one can use, for example, a hybridization technique using nucleic acids that
hybridize to these
genes as probes, or a gene amplification technique using DNA that hybridize to
the marker
genes as primers.
[000387] The probes or primers used for the testing can be designed based on
the nucleotide
sequences of the marker genes. The identification numbers for the nucleotide
sequences of
the respective marker genes are describer herein.
[000388] Further, it is to be understood that genes of higher animals
generally accompany
71

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
polymorphism in a high frequency. There are also many molecules that produce
isoforms
comprising mutually different amino acid sequences during the splicing
process. Any gene
associated with a cancer-related disease that has an activity similar to that
of a marker gene is
included in the marker genes, even if it has nucleotide sequence differences
due to
polymorphism or being an isoform.
[000389] It is also to be understood that the marker genes can include
homologs of other
species in addition to humans. Thus, unless otherwise specified, the
expression "marker
gene" refers to a homolog of the marker gene unique to the species or a
foreign marker gene
which has been introduced into an individual.
[000390] Also, it is to be understood that a "homolog of a marker gene" refers
to a gene derived
from a species other than a human, which can hybridize to the human marker
gene as a probe
under stringent conditions. Such stringent conditions are known to one skilled
in the art who
can select an appropriate condition to produce an equal stringency
experimentally or
empirically.
[000391] A polynucleotide comprising the nucleotide sequence of a marker gene
or a
nucleotide sequence that is complementary to the complementary strand of the
nucleotide
sequence of a marker gene and has at least 15 nucleotides, can be used as a
primer or probe.
Thus, a "complementary strand" means one strand of a double stranded DNA with
respect to
the other strand and which is composed of A:T (U for RNA) and G:C base pairs.
[000392] In addition, "complementary" means not only those that are completely
complementary to a region of at least 15 continuous nucleotides, but also
those that have a
nucleotide sequence homology of at least 40% in certain instances, 50% in
certain instances,
60% in certain instances, 70% in certain instances, at least 80%, 90%, and 95%
or higher.
The degree of homology between nucleotide sequences can be determined by an
algorithm,
BLAST, etc.
[000393] Such polynucleotides are useful as a probe to detect a marker gene,
or as a primer to
amplify a marker gene. When used as a primer, the polynucleotide comprises
usually 15 bp
to 100 bp, and in certain embodiments 15 bp to 35 bp of nucleotides. When used
as a probe,
a DNA comprises the whole nucleotide sequence of the marker gene (or the
complementary
strand thereof), or a partial sequence thereof that has at least 15 bp
nucleotides. When used
as a primer, the 3' region must be complementary to the marker gene, while the
5' region can
72

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
be linked to a restriction enzyme-recognition sequence or a tag.
[000394] "Polynucleotides" may be either DNA or RNA. These polynucleotides may
be either
synthetic or naturally-occurring. Also, DNA used as a probe for hybridization
is usually
labeled. Those skilled in the art readily understand such labeling methods.
Herein, the term
"oligonucleotide" means a polynucleotide with a relatively low degree of
polymerization.
Oligonucleotides are included in polynucleotides.
[000395] Tests for Cancer-related Diseases
[000396] Tests for a cancer-related disease using hybridization techniques can
be performed
using, for example, Northern hybridization, dot blot hybridization, or the DNA
microarray
technique. Furthermore, gene amplification techniques, such as the RT-PCR
method may be
used. By using the PCR amplification monitoring method during the gene
amplification step
in RT-PCR, one can achieve a more quantitative analysis of the expression of a
marker gene.
[000397] In the PCR gene amplification monitoring method, the detection target
(DNA or
reverse transcript of RNA) is hybridized to probes that are labeled with a
fluorescent dye and
a quencher which absorbs the fluorescence. When the PCR proceeds and Taq
polymerase
degrades the probe with its 5'-3' exonuclease activity, the fluorescent dye
and the quencher
draw away from each other and the fluorescence is detected. The fluorescence
is detected in
real time. By simultaneously measuring a standard sample in which the copy
number of a
target is known, it is possible to determine the copy number of the target in
the subject
sample with the cycle number where PCR amplification is linear. Also, one
skilled in the art
recognizes that the PCR amplification monitoring method can be carried out
using any
suitable method.
[000398] The method of testing for a cancer-related disease can be also
carried out by detecting
a protein encoded by a marker gene. Hereinafter, a protein encoded by a marker
gene is
described as a "marker protein." For such test methods, for example, the
Western blotting
method, the immunoprecipitation method, and the ELISA method may be employed
using an
antibody that binds to each marker protein.
[000399] Antibodies used in the detection that bind to the marker protein may
be produced by
any suitable technique. Also, in order to detect a marker protein, such an
antibody may be
appropriately labeled. Alternatively, instead of labeling the antibody, a
substance that
specifically binds to the antibody, for example, protein A or protein G, may
be labeled to
73

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
detect the marker protein indirectly. More specifically, such a detection
method can include
the ELISA method.
[000400] A protein or a partial peptide thereof used as an antigen may be
obtained, for
example, by inserting a marker gene or a portion thereof into an expression
vector,
introducing the construct into an appropriate host cell to produce a
transformant, culturing the
transformant to express the recombinant protein, and purifying the expressed
recombinant
protein from the culture or the culture supernatant. Alternatively, the amino
acid sequence
encoded by a gene or an oligopeptide comprising a portion of the amino acid
sequence
encoded by a full-length cDNA are chemically synthesized to be used as an
immunogen.
[000401] Furthermore, a test for a cancer-related disease can be performed
using as an index
not only the expression level of a marker gene but also the activity of a
marker protein in a
biological sample. Activity of a marker protein means the biological activity
intrinsic to the
protein. Various methods can be used for measuring the activity of each
protein.
[000402] Even if a patient is not diagnosed as being affected with a cancer-
related disease in a
routine test in spite of symptoms suggesting these diseases, whether or not
such a patient is
suffering from a cancer-related disease can be easily determined by performing
a test
according to the methods described herein.
[000403] More specifically, in certain embodiments, when the marker gene is
one of the genes
described herein, an increase or decrease in the expression level of the
marker gene in a
patient whose symptoms suggest at least a susceptibility to a cancer-related
disease indicates
that the symptoms are primarily caused by a cancer-related disease.
[000404] In addition, the tests are useful to determine whether a cancer-
related disease is
improving in a patient. In other words, the methods described herein can be
used to judge the
therapeutic effect of a treatment for a cancer-related disease. Furthermore,
when the marker
gene is one of the genes described herein, an increase or decrease in the
expression level of
the marker gene in a patient, who has been diagnosed as being affected by a
cancer-related
disease, implies that the disease has progressed more.
[000405] The severity and/or susceptibility to a cancer-related disease may
also be determined
based on the difference in expression levels. For example, when the marker
gene is one of
the genes described herein, the degree of increase in the expression level of
the marker gene
is correlated with the presence and/or severity of a cancer-related disease.
74

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000406] Control of Expression of Marker
[000407] In addition, the expression itself of a marker gene can be controlled
by introducing a
mutation(s) into the transcriptional regulatory region of the gene. Those
skilled in the art
understand such amino acid substitutions. Also, the number of amino acids that
are mutated
is not particularly restricted, as long as the activity is maintained.
Normally, it is within 50
amino acids, in certain non-limiting embodiments, within 30 amino acids,
within 10 amino
acids, or within 3 amino acids. The site of mutation may be any site, as long
as the activity is
maintained.
[000408] Screening Methods
[000409] In yet another aspect, there is provided herein screening methods for
candidate
compounds for therapeutic agents to treat a cancer-related disease. One or
more marker
genes are selected from the group of genes described herein. A therapeutic
agent for a
cancer-related disease can be obtained by selecting a compound capable of
increasing or
decreasing the expression level of the marker gene(s).
[000410] It is to be understood that the expression "a compound that increases
the expression
level of a gene" refers to a compound that promotes any one of the steps of
gene
transcription, gene translation, or expression of a protein activity. On the
other hand, the
expression "a compound that decreases the expression level of a gene", as used
herein, refers
to a compound that inhibits any one of these steps.
[000411] In particular aspects, the method of screening for a therapeutic
agent for a cancer-
related disease can be carried out either in vivo or in vitro. This screening
method can be
performed, for example, by (1) administering a candidate compound to an animal
subject; (2)
measuring the expression level of a marker gene(s) in a biological sample from
the animal
subject; or (3) selecting a compound that increases or decreases the
expression level of a
marker gene(s) as compared to that in a control with which the candidate
compound has not
been contacted.
[000412] In still another aspect, there is provided herein a method to assess
the efficacy of a
candidate compound for a pharmaceutical agent on the expression level of a
marker gene(s)
by contacting an animal subject with the candidate compound and monitoring the
effect of
the compound on the expression level of the marker gene(s) in a biological
sample derived
from the animal subject. The variation in the expression level of the marker
gene(s) in a

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
biological sample derived from the animal subject can be monitored using the
same technique
as used in the testing method described above. Furthermore, based on the
evaluation, a
candidate compound for a pharmaceutical agent can be selected by screening.
[000413] Kits
[000414] In another aspect, there is provided various diagnostic and test
kits. In one
embodiment, a kit is useful for assessing whether a patient is afflicted with
a cancer-related
disease. The kit comprises a reagent for assessing expression of a marker. In
another
embodiment, a kit is useful for assessing the suitability of a chemical or
biologic agent for
inhibiting a cancer-related disease in a patient. Such a kit comprises a
reagent for assessing
expression of a marker, and may also comprise one or more of such agents.
[000415] In a further embodiment, the kits are useful for assessing the
presence of cancer-
related disease cells or treating cancer-related diseases. Such kits comprise
an antibody, an
antibody derivative or an antibody fragment, which binds specifically with a
marker protein
or a fragment of the protein. Such kits may also comprise a plurality of
antibodies, antibody
derivatives or antibody fragments wherein the plurality of such antibody
agents binds
specifically with a marker protein or a fragment of the protein.
[000416] In an additional embodiment, the kits are useful for assessing the
presence of cancer-
related disease cells, wherein the kit comprises a nucleic acid probe that
binds specifically
with a marker nucleic acid or a fragment of the nucleic acid. The kit may also
comprise a
plurality of probes, wherein each of the probes binds specifically with a
marker nucleic acid,
or a fragment of the nucleic acid.
[000417] The compositions, kits and methods described herein can have the
following uses,
among others: 1) assessing whether a patient is afflicted with a cancer-
related disease; 2)
assessing the stage of a cancer-related disease in a human patient; 3)
assessing the grade of a
cancer-related disease in a patient; 4) assessing the nature of a cancer-
related disease in a
patient; 5) assessing the potential to develop a cancer-related disease in a
patient; 6) assessing
the histological type of cells associated with a cancer-related disease in a
patient; 7) making
antibodies, antibody fragments or antibody derivatives that are useful for
treating a cancer-
related disease and/or assessing whether a patient is afflicted with a cancer-
related disease; 8)
assessing the presence of cancer-related disease cells; 9) assessing the
efficacy of one or more
test compounds for inhibiting a cancer-related disease in a patient; 10)
assessing the efficacy
76

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
of a therapy for inhibiting a cancer-related disease in a patient; 11)
monitoring the
progression of a cancer-related disease in a patient; 12) selecting a
composition or therapy for
inhibiting a cancer-related disease in a patient; 13) treating a patient
afflicted with a cancer-
related disease; 14) inhibiting a cancer-related disease in a patient; 15)
assessing the harmful
potential of a test compound; and 16) preventing the onset of a cancer-related
disease in a
patient at risk for developing a cancer-related disease.
[000418] The kits are useful for assessing the presence of cancer-related
disease cells (e.g. in a
sample such as a patient sample). The kit comprises a plurality of reagents,
each of which is
capable of binding specifically with a marker nucleic acid or protein.
Suitable reagents for
binding with a marker protein include antibodies, antibody derivatives,
antibody fragments,
and the like. Suitable reagents for binding with a marker nucleic acid (e.g. a
genomic DNA,
an MRNA, a spliced MRNA, a cDNA, or the like) include complementary nucleic
acids. For
example, the nucleic acid reagents may include oligonucleotides (labeled or
non-labeled)
fixed to a substrate, labeled oligonucleotides not bound with a substrate,
pairs of PCR
primers, molecular beacon probes, and the like.
[000419] The kits may optionally comprise additional components useful for
performing the
methods described herein. By way of example, the kit may comprise fluids (e.g.
SSC buffer)
suitable for annealing complementary nucleic acids or for binding an antibody
with a protein
with which it specifically binds, one or more sample compartments, an
instructional material
which describes performance of the method, a sample of normal cells, a sample
of cancer-
related disease cells, and the like.
[000420]
[000421] Animal Model
[000422] Non-human animal model can be produced for assessment of at least one
cancer-
related disease. The method includes exposing the animal to repeated doses of
at least one
chemical believed to cause the cancer if interest. In certain aspects, the
method further
includes collecting one or more selected samples from the animal; and
comparing the
collected sample to one or more indicia of potential cancer initiation or
development.
[000423] A method of producing the animal model includes: maintaining the
animal in a
specific chemical-free environment and sensitizing the animal with at least
one chemical
believed to cause the cancer. In certain embodiments, at least a part of the
animal is
77

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
sensitized by multiple sequential exposures.
[000424] A method of screening for an agent for effectiveness against at least
one cancer-
related disease generally includes: administering at least one agent to a test
animal,
determining whether the agent reduces or aggravates one or more symptoms of
the cancer-
related disease; correlating a reduction in one or more symptoms with
effectiveness of the
agent against the cancer-related disease; or correlating a lack of reduction
in one or more
symptoms with ineffectiveness of the agent. The animal model is useful for
assessing one or
more metabolic pathways that contribute to at least one of initiation,
progression, severity,
pathology, aggressiveness, grade, activity, disability, mortality, morbidity,
disease sub-
classification or other underlying pathogenic or pathological feature of at
least one cancer-
related disease. The analysis can be by one or more of: hierarchical
clustering, signature
network construction, mass spectroscopy proteomic analysis, surface plasmon
resonance,
linear statistical modeling, partial least squares discriminant analysis, and
multiple linear
regression analysis.
[000425] The animal model can be assessed for at least one cancer-related
disease, by
examining an expression level of one or more markers, or a functional
equivalent thereto.
[000426] The animal models can be used for the screening of therapeutic agents
useful for
treating or preventing a cancer-related disease. Accordingly, the methods are
useful for
identifying therapeutic agents for treating or preventing a cancer-related
disease. The
methods comprise administering a candidate agent to an animal model made by
the methods
described herein, assessing at least one cancer-related disease response in
the animal model
as compared to a control animal model to which the candidate agent has not
been
administered. If at least one cancer-related disease response is reduced in
symptoms or
delayed in onset, the candidate agent is an agent for treating or preventing
the cancer-related
disease.
[000427] The animal models for a cancer-related disease can include an animal
where the
expression level of one or more marker genes or a gene functionally equivalent
to the marker
gene has been elevated in the animal model. A "functionally equivalent gene"
as used herein
generally is a gene that encodes a protein having an activity similar to a
known activity of a
protein encoded by the marker gene. A representative example of a functionally
equivalent
gene includes a counterpart of a marker gene of a subject animal, which is
intrinsic to the
animal.
78

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[000428] The animal model for a cancer-related disease is useful for detecting
physiological
changes due to a cancer-related disease. In certain embodiments, the animal
model is useful
to reveal additional functions of marker genes and to evaluate drugs whose
targets are the
marker genes.
[000429] In one embodiment, an animal model for a cancer-related disease can
be created by
controlling the expression level of a counterpart gene or administering a
counterpart gene.
The method can include creating an animal model for a cancer-related disease
by controlling
the expression level of a gene selected from the group of genes described
herein. In another
embodiment, the method can include creating an animal model for a cancer-
related disease by
administering the protein encoded by a gene described herein, or administering
an antibody
against the protein. It is to be also understood, that in certain other
embodiments, the marker
can be over-expressed such that the marker can then be measured using
appropriate methods.
[000430] In another embodiment, an animal model for a cancer-related disease
can be created
by introducing a gene selected from such groups of genes, or by administering
a protein
encoded by such a gene.
[000431] In another embodiment, a cancer-related disease can be induced by
suppressing the
expression of a gene selected from such groups of genes or the activity of a
protein encoded
by such a gene. An antisense nucleic acid, a ribozyme, or an RNAi can be used
to suppress
the expression. The activity of a protein can be controlled effectively by
administering a
substance that inhibits the activity, such as an antibody.
[000432] The animal model is useful to elucidate the mechanism underlying a
cancer-related
disease and also to test the safety of compounds obtained by screening. For
example, when
an animal model develops the symptoms of a cancer-related disease, or when a
measured
value involved in a certain a cancer-related disease alters in the animal, a
screening system
can be constructed to explore compounds having activity to alleviate the
disease.
[000433] As used herein, the expression "an increase in the expression level"
refers to any one
of the following: where a marker gene introduced as a foreign gene is
expressed artificially;
where the transcription of a marker gene intrinsic to the subject animal and
the translation
thereof into the protein are enhanced; or where the hydrolysis of the protein,
which is the
translation product, is suppressed. As used herein, the expression "a decrease
in the
expression level" refers to either the state in which the transcription of a
marker gene of the
79

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
subject animal and the translation thereof into the protein are inhibited, or
the state in which
the hydrolysis of the protein, which is the translation product, is enhanced.
The expression
level of a gene can be determined, for example, by a difference in signal
intensity on a DNA
chip. Furthermore, the activity of the translation product--the protein--can
be determined by
comparing with that in the normal state.
[000434] It is also within the contemplated scope that the animal model can
include transgenic
animals, including, for example animals where a marker gene has been
introduced and
expressed artificially; marker gene knockout animals; and knock-in animals in
which another
gene has been substituted for a marker gene. A transgenic animal, into which
an antisense
nucleic acid of a marker gene, a ribozyme, a polynucleotide having an RNAi
effect, or a
DNA functioning as a decoy nucleic acid or such has been introduced, can be
used as the
transgenic animal. Such transgenic animals also include, for example, animals
in which the
activity of a marker protein has been enhanced or suppressed by introducing a
mutation(s)
into the coding region of the gene, or the amino acid sequence has been
modified to become
resistant or susceptible to hydrolysis. Mutations in an amino acid sequence
include
substitutions, deletions, insertions, and additions.
THERAPEUTIC APPLICATIONS
[0227] The invention is widely applicable to a variety of situations where it
is
desirable to be able to regulate the level of gene expression, such as by
turning gene
expression "on" and "off", in a rapid, efficient and controlled manner without
causing
pleiotropic effects or cytotoxicity. The invention may be particularly useful
for gene
therapy purposes in humans, in treatments for either genetic or acquired
diseases. The
general approach of gene therapy involves the introduction of one or more
nucleic
acid molecules into cells such that one or more gene products encoded by the
introduced genetic material are produced in the cells to restore or enhance a
functional
activity. For reviews on gene therapy approaches Anderson, et al. (1992;
Miller et al.
(1992); Friedmann et al. (1989); and Cournoyer et al. (1990). However, current
gene
therapy vectors typically utilize constitutive regulatory elements which are
responsive
to endogenous transcriptions factors. These vector systems do not allow for
the ability

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
to modulate the level of gene expression in a subject. In contrast, the
regulatory
system of the invention provides this ability.
[0228] To use the system of the invention for gene therapy purposes, at least
one DNA
molecule is introduced into cells of a subject in need of gene therapy (e.g.,
a human
subject suffering from a genetic or acquired disease) to modify the cells. The
cells are
modified to comprise: 1) nucleic acid encoding an inducible regulator of the
invention
in a form suitable for expression of the inducible regulator in the host
cells; and 2) an
siRNA (e.g., for therapeutic purposes) operatively linked to a tissue-specific
promoter
such as an s-shipl promoter. A single DNA molecule encoding components of the
regulatory system of the invention can be used, or alternatively, separate DNA
molecules encoding each component can be used. The cells of the subject can be
modified ex vivo and then introduced into the subject or the cells can be
directly
modified in vivo by conventional techniques for introducing nucleic acid into
cells.
Thus, the regulatory system of the invention offers the advantage over
constitutive
regulatory systems of allowing for modulation of the level of gene expression
depending upon the requirements of the therapeutic situation.
[0229] Genes of particular interest to be knocked down or knocked out in cells
of a
subject for treatment of genetic or acquired diseases include those encoding a
deleterious gene product, such as an abnormal protein. Examples of non-
limiting
specific diseases include anemia, blood-related cancers, Parkinson's disease,
and
diabetes.
[0230] The present invention can be applied to develop autologous or
allogeneic cell
lines for therapeutical purposes. For example, gene therapy applications of
particular
interest in cell and/or organ transplantation are utilized with the present
invention. In
exemplary embodiments, downregulation of transplantation antigens (such as,
for
example, by downregulation of beta2-microglobulin expression via siRNA) allows
for
transplantation of allogeneic cells while minimizing the risk of rejection by
the
patient's immune system. The present invention would allow for a switch off of
the
81

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
RNAi in case of adverse effects (e.g. uncontrollable replication of the
transplanted
cells).
[0231] Cells types that can be subjected to the present invention include
hematopoietic
stem cells, myoblasts, hepatocytes, lymphocytes, airway epithelium, skin
epithelium,
islets, dopaminergic neurons, keratinocytes, and so forth. For further
descriptions of
cell types, genes and methods for gene therapy see e.g., Armentano et al.
(1990);
Wolff et al. (1990); Chowdhury et al. (1991); Ferry et al. (1991); Quantin et
al (1992);
Dai et al. (1992); van Beusechem et al. (1992); Rosenfeld et al. (1992); Kay
et al.
(1992); Cristiano et al (1993); Hwu et al (1993); and Herz and Gerard (1993).
[0232] In particular embodiments of the present invention, there is a method
of
treating any disease condition amenable to treatment with an s-ship promoter.
In
specific embodiments, the method comprises preparing a polynucleotide
construct
having a region encoding a therapeutic or diagnostic (marker) gene that is
operably
linked to a promoter, wherein the gene encoded by the construct is for the
treatment of
the disease condition.
[0233] A. Pharmaceutical Formulations, Delivery, and Treatment Regimens
[0234] In an embodiment of the present invention, methods of treatment are
contemplated. An effective amount of the pharmaceutical composition,
generally, is
defined as that amount sufficient to detectably and repeatedly to ameliorate,
reduce,
minimize or limit the extent of the disease or its symptoms. More rigorous
definitions
may apply, including elimination, eradication or cure of disease.
[0235] The routes of administration will vary, naturally, with the location
and nature
of the lesion, and include, e.g., intradermal, transdermal, parenteral,
intravenous,
intramuscular, intranasal, subcutaneous, percutaneous, intratracheal,
intraperitoneal,
intratumoral, perfusion, lavage, direct injection, and oral administration and
formulation.
82

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[0236] Solutions of the active compounds as free base or pharmacologically
acceptable salts may be prepared in water suitably mixed with a surfactant,
such as
hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid
polyethylene glycols, and mixtures thereof and in oils. Under ordinary
conditions of
storage and use, these preparations contain a preservative to prevent the
growth of
microorganisms. The pharmaceutical forms suitable for injectable use include
sterile
aqueous solutions or dispersions and sterile powders for the extemporaneous
preparation of sterile injectable solutions or dispersions (U.S. Pat. No.
5,466,468,
specifically incorporated herein by reference in its entirety). In all cases
the form must
be sterile and must be fluid to the extent that easy syringability exists. It
must be
stable under the conditions of manufacture and storage and must be preserved
against
the contaminating action of microorganisms, such as bacteria and fungi. The
carrier
can be a solvent or dispersion medium containing, for example, water, ethanol,
polyol
(e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the
like), suitable
mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained,
for
example, by the use of a coating, such as lecithin, by the maintenance of the
required
particle size in the case of dispersion and by the use of surfactants. The
prevention of
the action of microorganisms can be brought about by various antibacterial and
antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid,
thimerosal, and the like. In many cases, it will be preferable to include
isotonic agents,
for example, sugars or sodium chloride. Prolonged absorption of the injectable
compositions can be brought about by the use in the compositions of agents
delaying
absorption, for example, aluminum monostearate and gelatin.
[0237] For parenteral administration in an aqueous solution, for example, the
solution
should be suitably buffered if necessary and the liquid diluent first rendered
isotonic
with sufficient saline or glucose. These particular aqueous solutions are
especially
suitable for intravenous, intramuscular, subcutaneous, intratumoral and
intraperitoneal
administration. In this connection, sterile aqueous media that can be employed
will be
known to those of skill in the art in light of the present disclosure. For
example, one
83

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
dosage may be dissolved in 1 ml of isotonic NaC1 solution and either added to
1000
ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see
for
example, "Remington's Pharmaceutical Sciences" 15th Edition, pages 1035-1038
and
1570-1580). Some variation in dosage will necessarily occur depending on the
condition of the subject being treated. The person responsible for
administration will,
in any event, determine the appropriate dose for the individual subject.
Moreover, for
human administration, preparations should meet sterility, pyrogenicity,
general safety
and purity standards as required by FDA Office of Biologics standards.
[0238] Sterile injectable solutions are prepared by incorporating the active
compounds
in the required amount in the appropriate solvent with various of the other
ingredients
enumerated above, as required, followed by filtered sterilization. Generally,
dispersions are prepared by incorporating the various sterilized active
ingredients into
a sterile vehicle which contains the basic dispersion medium and the required
other
ingredients from those enumerated above. In the case of sterile powders for
the
preparation of sterile injectable solutions, the preferred methods of
preparation are
vacuum-drying and freeze-drying techniques which yield a powder of the active
ingredient plus any additional desired ingredient from a previously sterile-
filtered
solution thereof.
[0239] The compositions disclosed herein may be formulated in a neutral or
salt form.
Pharmaceutically-acceptable salts, include the acid addition salts (formed
with the free
amino groups of the protein) and which are formed with inorganic acids such
as, for
example, hydrochloric or phosphoric acids, or such organic acids as acetic,
oxalic,
tartaric, mandelic, and the like. Salts formed with the free carboxyl groups
can also be
derived from inorganic bases such as, for example, sodium, potassium,
ammonium,
calcium, or ferric hydroxides, and such organic bases as isopropylamine,
trimethylamine, histidine, procaine and the like. Upon formulation, solutions
will be
administered in a manner compatible with the dosage formulation and in such
amount
as is therapeutically effective. The formulations are easily administered in a
variety of
dosage forms such as injectable solutions, drug release capsules and the like.
84

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
[0240] As used herein, "carrier" includes any and all solvents, dispersion
media,
vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic
and
absorption delaying agents, buffers, carrier solutions, suspensions, colloids,
and the
like. The use of such media and agents for pharmaceutical active substances is
well
known in the art. Except insofar as any conventional media or agent is
incompatible
with the active ingredient, its use in the therapeutic compositions is
contemplated.
Supplementary active ingredients can also be incorporated into the
compositions.
[0241] The phrase "pharmaceutically-acceptable" or "pharmacologically-
acceptable"
refers to molecular entities and compositions that do not produce an allergic
or similar
untoward reaction when administered to a human. The preparation of an aqueous
composition that contains a protein as an active ingredient is well understood
in the
art. Typically, such compositions are prepared as injectables, either as
liquid solutions
or suspensions; solid forms suitable for solution in, or suspension in, liquid
prior to
injection can also be prepared.
[0242] B. Combination Treatments
[0243] The compounds and methods of the present invention may be used in the
context of traditional therapies. In order to increase the effectiveness of a
treatment
with the compositions of the present invention, it may be desirable to combine
these
compositions with other agents effective in the treatment of those diseases
and
conditions. For example, the treatment of a cancer may be implemented with
therapeutic compounds of the present invention and other anti-cancer
therapies, such
as anti-cancer agents or surgery. Likewise, the treatment of a vascular
disease or
condition may involve both the present invention and conventional vascular
agents or
therapies.
[0244] Various combinations may be employed; for example, a host cell of the
present
invention is "A" and the secondary anti-cancer agent/therapy is "B": TABLE-US-

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
00005 A/B/A B/A/B B/B/A A/A/B A/B/B B/A/A A/B/B/B B/A/B/B B/B/B/A
B/B/A/B A/A/B/B A/B/A/B A/B/B/A B/B/A/A B/A/B/A B/A/A/B A/A/A/B B/A/A/A
A/B/A/A A/A/B/A
[0245] Administration of the therapeutic expression constructs of the present
invention to a patient will follow general protocols for the administration of
that
particular secondary therapy, taking into account the toxicity, if any, of the
treatment.
It is expected that the treatment cycles would be repeated as necessary. It
also is
contemplated that various standard therapies, as well as surgical
intervention, may be
applied in combination with the described therapy.
[000435] All patents, patent applications and references cited herein are
incorporated in their
entirety by reference. While the invention has been described and exemplified
in sufficient
detail for those skilled in this art to make and use it, various alternatives,
modifications and
improvements should be apparent without departing from the spirit and scope of
the
invention. One skilled in the art readily appreciates that the present
invention is well adapted
to carry out the objects and obtain the ends and advantages mentioned, as well
as those
inherent therein.
[000436] The methods and reagents described herein are representative of
preferred
embodiments, are exemplary, and are not intended as limitations on the scope
of the
invention. Modifications therein and other uses will occur to those skilled in
the art. These
modifications are encompassed within the spirit of the invention and are
defined by the scope
of the claims. It will also be readily apparent to a person skilled in the art
that varying
substitutions and modifications may be made to the invention disclosed herein
without
departing from the scope and spirit of the invention.
[000437] It should be understood that although the present invention has been
specifically
disclosed by preferred embodiments and optional features, modifications and
variations of the
concepts herein disclosed may be resorted to by those skilled in the art, and
that such
modifications and variations are considered to be within the scope of this
invention as defined
by the appended claims.
[000438] While the invention has been described with reference to various and
preferred
86

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
embodiments, it should be understood by those skilled in the art that various
changes may be
made and equivalents may be substituted for elements thereof without departing
from the
essential scope of the invention. In addition, many modifications may be made
to adapt a
particular situation or material to the teachings of the invention without
departing from the
essential scope thereof.
[000439] Therefore, it is intended that the invention not be limited to the
particular embodiment
disclosed herein contemplated for carrying out this invention, but that the
invention will
include all embodiments falling within the scope of the claims.
[000440] All of the compositions and/or methods and/or apparatus disclosed and
claimed herein
can be made and executed without undue experimentation in light of the present
disclosure.
While the compositions and methods of this invention have been described in
terms of
preferred embodiments, it will be apparent to those of skill in the art that
variations may be
applied to the compositions and/or methods and/or apparatus and in the steps
or in the
sequence of steps of the method described herein without departing from the
concept, spirit
and scope of the invention. More specifically, it will be apparent that
certain agents that are
both chemically and physiologically related may be substituted for the agents
described
herein while the same or similar results would be achieved. All such similar
substitutes and
modifications apparent to those skilled in the art are deemed to be within the
spirit, scope and
concept of the invention as defined by the appended claims.
[000441] Throughout this disclosure, various publications, patents and
published patent
specifications are referenced by an identifying citation. The disclosures of
these publications,
patents and published patent specifications are hereby incorporated by
reference into the
present disclosure to more fully describe the state of the art to which this
invention pertains.
[000442] Unless otherwise defined, all technical and scientific terms used
herein have the same
meaning as commonly understood by one of ordinary skill in the art (e.g., in
cell culture,
molecular genetics, nucleic acid chemistry, hybridization techniques and
biochemistry).
Standard techniques are used for molecular, genetic and biochemical methods
which are
within the skill of the art. Such techniques are explained fully in the
literature. See, for
example, Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook,
Fritsch and
Maniatis (Cold Spring Harbor Laboratory Press: 1989); DNA Cloning, Volumes I
and II
(Glover ed., 1985); Oligonucleotide Synthesis (Gait ed., 1984); Mullis et al.
U.S. Pat. No:
4,683,195; Nucleic Acid Hybridization (Hames & Higgins eds., 1984);
Transcription And
87

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
Translation (Hames & Higgins eds., 1984); Culture Of Animal Cells (R. I.
Freshney, Alan R.
Liss, Inc., 1987); Immobilized Cells And Enzymes (IRL Press, 1986); Perbal, A
Practical
Guide To Molecular Cloning (1984); the treatise, Methods In Enzymology
(Academic Press,
Inc., N.Y.); Gene Transfer Vectors For Mammalian Cells (Miller and Calos eds.,
1987, Cold
Spring Harbor Laboratory); Methods In Enzymology, Vols. 154 and 155 (Wu et al.
eds.),
Immunochemical Methods In Cell And Molecular Biology (Mayer and Walker, eds.,
Academic Press, London, 1987); Handbook Of Experimental Immunology, Volumes I-
IV
(Weir and Blackwell, eds., 1986); The Laboratory Rat, editor in chief: Mark A.
Suckow;
authors: Sharp and LaRegina. CRC Press, Boston, 1988, which are incorporated
herein by
reference) and chemical methods.
[000443] REFERENCES
[000444] The publication and other material used herein to illuminate the
invention or provide
additional details respecting the practice of the invention, are incorporated
be reference
herein, and for convenience are provided in the following bibliography.
[000445] Citation of the any of the documents recited herein is not intended
as an admission
that any of the foregoing is pertinent prior art. All statements as to the
date or representation
as to the contents of these documents is based on the information available to
the applicant
and does not constitute any admission as to the correctness of the dates or
contents of these
documents.
1. Ambros, V. (2003). MicroRNA pathways in flies and worms: growth, death,
fat,
stress, and timing. Cell 113, 673-676.
2. Ambros, V. (2004). The functions of animal microRNAs. Nature 431, 350-355.
3. Balmain, A., Gray, J. W., and Ponder, B. A. (2003). The genetics and
genomics of
cancer. Nat Genet 33 Suppl, 238-244.
4. Bejerano, G., Haussler, D., and Blanchette, M. (2004a). Into the heart of
darkness:
large-scale clustering of human non-coding DNA. Bioinformatics 20, 140-148.
5. Bejerano, G., Pheasant, M., Makunin, I., Stephen, S., Kent, W. J., Mattick,
J. S., and
Haussler, D.
6. (2004b). Ultraconserved elements in the human genome. Science 304, 1321-
1325.
Benjamini Y and Hochberg Y. Controlling the False Discovery Rate: a Practical
and
Powerful
7. Approach to Multiple Testing. Journal of the Royal Statistical Society. B,
57, 289-
300. Berezikov, E. and Plasterk, R. H. Camels and zebrafish, viruses and
cancer: a
microRNA update
8. (2005a). Hum Mol Genet. 14, R183-90.
9. Berezikov, E., Guryev, V., van de Belt, J., Wienholds, E., Plasterk, R. H.,
and
Cuppen, E. (2005b). Phylogenetic shadowing and computational identification of
human
microRNA genes. Cell 120, 21-24.
88

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
10. Bejerano, G., Lowe, C. B., Ahituv, N., King, B., Siepel, A., Salama, S.
R., Rubin, E.
M., Kent, W. J., and Haussler, D. (2006).A distal enhancer and an
ultraconserved exon are
derived from a novel retroposon. Nature 441, 87-90.
11. Bishop, J. M. (1991). Molecular themes in oncogenesis. Cell 64, 235-248.
12. Calin, G. A., Dumitru, C. D., Shimizu, M., Bichi, R., Zupo, S., Noch, E.,
Aldler, H.,
Rattan, S., Keating, M., Rai, K., et al. (2002). Frequent deletions and down-
regulation of
microRNA genes riR15 and miR16 at 13g14 in chronic lymphocytic leukemia. Proc
Natl
Acad Sc U S A 99, 15524-15529.
13. Calin, G. A., Liu, C. G., Sevignani, C., Ferracin, M., Felli, N., Dumitru,
C. D.,
Shimizu, M., Cimmino, A., Zupo, S., Dono, M., et al. (2004a). MicroRNA
profiling reveals
distinct signatures in B cell chronic lymphocytic leukemias. Proc Natl Acad
Sci U S A 101,
11755-11760.
14. Calin, G. A., Sevignani, C., Dumitru, C. D., Hyslop, T., Noch, E.,
Yendamuri, S.,
Shimizu, M., Rattan, S., Bullrich, F., Negrini; M., and Croce, C. M. (2004b).
Human
microRNA genes are frequently located at fragile sites and genomic regions
involved in
cancers. Proc Natl Acad Sci U S A 101, 2999-3004.
15. Calin, G. A., Ferracin, M., Cimmino, A., Di Leva, G., Shimizu, M., Wojcik,
S., Iorio,
M. V., Visone, R., Sever, N. I., Fabbri, M., et al. (2005a). A Unique MicroRNA
Signature
Associated with Prognostic Factors and Disease Progression in B cell Chronic
Lymphocytic
Leukemia. N Engl J Med 352, 1667-1676.
16. Calin, G. A., and Croce, C. M. (2006a). MicroRNA signatures in human
cancers.
Nature Reviews Cancer 6, 857-866.
17. Calin, G. A., and Croce, C. M. (2006b). MicroRNA-cancer connection: the
beginning
of a new tale. Cancer Res 66, 7390-7394.
18. Chen, C., Ridzon, D. A., Broomer, A. J., Zhou, Z., Lee, D. H., Nguyen, J.
T.,
Barbisin, M., Xu, N. L., Mahuvakar, V. R., Andersen, M. R., et al. (2005).
Real-time
quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res 33, e179.
19. Chiorazzi, N., Rai, K. R., and Ferrarini, M. (2005). Chronic lymphocytic
leukemia. N
Engl J Med 352, 804-815.
20. Cimmino, A., Calin, G. A., Fabbri, M., Iorio, M. V., Ferracin, M.,
Shimizu, M.,
Wojcik, S. E., Aqeilan, R., Zupo, S., Dono, M., et al. (2005). miR-15 and miR-
16 induce
apoptosis by targeting BCL2. Proc Natl Acad Sc U S A 102, 13944-13949.
21. Croce, C. M., and Calin, G. A. (2005). miRNAs, Cancer, and Stem Cell
Division. Cell
122, 6-7. de la Chapelle, A. (2004). Genetic predisposition to colorectal
cancer. Nature
Reviews Cancer 4, 769-780.
22. Dermitzakis, E. T., Reymond, A., and Antonarakis, S. E. (2005). Conserved
non-
genic sequences - an unexpected feature of mammalian genomes. Nat Rev Genet 6,
151-157.
23. Duret, L., Dorkeld, F., and Gautier, C. (1993). Strong conservation of non-
coding
sequences during vertebrates evolution: potential involvement in post-
transcriptional
regulation of gene expression. Nucleic Acids Res 21, 2315-2322.
24. Eis, P. S., Tam, W., Sun, L., Chadburn, A., Li, Z., Gomez, M. F., Lund,
E., and
Dahlberg, J. E. (2005). Accumulation of miR-155 and BIC RNA in human B cell
lymphomas. Proc Natl Acad Sci U S A 102, 3627-3632.
25. Esquela-Kerscher, A., and Slack, F. J. (2006). Oncomirs - microRNAs with a
role in
cancer. Nat Rev Cancer 6, 259-269.
26. Furuno, M., Pang, K. C., Ninomiya, N., Fukuda, S., Frith, M. C., Bult, C.,
Kai, C.,
Kawai, J., Carninci, P., Hayashizaki, Y., et al. (2006). Clusters of
internally primed
transcripts reveal novel long noncoding RNAs. PLoS Genet 2, e37.
27. Hunter, T. (1991). Cooperation between oncogenes. Cell 64, 249-270.
89

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
28. Huntley, S., Baggott, D. M., Hamilton, A. T., Tran-Gyamfi, M., Yang, S.
C., Kim, J.,
Gordon, L., Branscomb, E., and Stubbs, S. (2006). A comprehensive catalog of
human
KRAB-associated zinc finger genes: Insights into the evolutionary history of a
large family of
transcriptional repressors. Genome Res 16:669-677.
29. Gaur A, Jewell DA, Liang Y, Ridzon D, Moore JH, Chen C, Ambros VR, Israel
MA.
Characterization of MicroRNA Expression Levels and Their Biological Correlates
in Human
Cancer Cell Lines. Cancer Res. 2007 Mar 15;67(6):2456-68.
30. Iorio, M. V., Ferracin, M., Liu, C. G., Veronese, A., Spizzo, R.,
Sabbioni, S., Magri,
E., Pedriali, M., Fabbri, M., Campiglio, M., et al. (2005). microRNA gene
expression
deregulation in human breast cancer. Cancer Res 65, 7065-7070.
31. Ji, P., Diederichs, S., Wang, W., Boing, S., Metzger, R., Schneider, P.
M., Tidow, N.,
Brandt, B., Buerger, H., Bulk, E., et al. (2003). MALAT-1, a novel noncoding
RNA, and
thymosin beta4 predict metastasis and survival in early-stage non-small cell
lung cancer.
Oncogene 22, 8031-8041.
32. Kluiver, J., Poppema, S., de Jong, D., Blokzijl, T., Harms, G., Jacobs,
S., Kroesen, B.
J., and van den Berg, A. (2005). BIC and miR-155 are highly expressed in
Hodgkin, primary
mediastinal and diffuse large B cell lymphomas. J Patho1207, 243-249.
33. Liu, C.-G., Cahn, G. A., Meloon, B., Gamliel, N., Sevignani, C., Ferracin,
M.,
Dumitru, D. C., Shimizu, M., Zupo, S., Dono, M., et al. (2004). An
oligonucleotide
microchip for genome-wide miRNA profiling in human and mouse tissues. Proc
Natl Acad
Sci U S A 101, 9740-9744.
34. Lu, J., Getz, G., Miska, E. A., Alvarez-Saavedra, E., Lamb, J., Peck, D.,
Sweet-
Cordero, A., Ebert, B. L., Mak, R. H., Ferrando, A. A., et al. (2005).
MicroRNA expression
profiles classify human cancers. Nature 435, 834-838.
35. Meisler, M. H. (2001). Evolutionarily conserved noncoding DNA in the human
genome: how much and what for? Genome Res 11, 1617-1618.
36. Ng, D., Toure, 0., Wei, M.H., Arthur, D.C., Abbasi, F., Fontaine, L.,
Marti, G.E.,
Fraumeni, J.F.J., Goldin, L.R., Caporaso, N.E., and Toro, J.R. (2007).
Identification of a
novel chromosome region, 13g21.33-g22.2, for susceptibility genes in familial
chronic
lyinphocytic leukemia. Blood 109, 916-925.
37. Nobrega, M. A., Ovcharenko, I., Afzal, V., and Rubin, E. M. (2003).
Scanning human
gene deserts for long-range enhancers. Science 302, 413.
38. Nobrega, M. A., Zhu, Y., Plajzer-Frick, I., Afzal, V., and Rubin, E. M.
(2004).
Megabase deletions of gene deserts result in viable mice. Nature 431, 988-993.
39. Pennacchio, L. A., Ahituv, N., Moses, A. M., Prabhakar, S., Nobrega, M.
A.,
Shoukry, M., Minovitsky, S., Dubchak, I., Holt, A., Lewis, K. D., et al.
(2006). In vivo
enhancer analysis of human conserved non-coding sequences. Nature Nov 5; [Epub
ahead of
print].
40. Raveche ES, Salerno E, Scaglione BJ, Manohar V, Abbasi F, Lin YC,
Fredrickson T,
Landgraf P, Ramachandra S, Huppi K, Toro JR, Zenger VE, Metcalf RA, Marti GE.
Abnormal microRNA-161ocus with synteny to human 13814 linked to CLL in NZB
mice.
Blood. 2007 Mar 9; [Epub ahead of print]
41. Reis, E. M., Louro, R., Nakaya, H. I., and Verjovski-Almeida, S. (2005).
As antisense
RNA gets intronic. OMICS 9.
42. Rhodes, D. R., and Chinnaiyan, A. M. (2005). Integrative analysis of the
cancer
transcriptome. Nat Genet 37, Supp1:S31-37.
43. Rigoutsos, I., Huynh, T., Miranda, K., Tsirigos, A., McHardy, A., and
Platt, D.
(2006). Short blocks from the noncoding parts of the human genome have
instances within
nearly all known genes and relate to biological processes. Proc Natl Acad Sci
U S A 103,
66056610.

CA 02695514 2010-02-03
WO 2009/020905 PCT/US2008/072081
44. Schmittgen, T. D., Jiang, J., Liu, Q., and yang, L. (2004). A high-
throughput method
to monitor the expression of microRNA precursor. Nucleic Acid Research 32, 43-
53.
45. Thomas, J. W., Touchman, J. W., Blakesley, R. W., Bouffard, G. G.,
Beckstrom-
Sternberg, S. M., Margulies, E. H., Blanchette, M., Siepel, A. C., Thomas, P.
J., McDowell, J.
C., et al. (2003). Comparative analyses of multi-species sequences from
targeted genomic
regions. Nature 424, 788-793.
46. Volinia, S., Calin, G. A., Liu, C.-G., Ambs, S., Cimmino, A., Petrocca,
F., Visone, R.,
Iorio, M.
47. V., Roldo, C., Ferracin, M., et al. (2006). A microRNA expression
signature of human
48. solid tumors define cancer gene targets. Proc Natl Acad Sc U S A 103, 2257-
2261.
Weinberg, R. A. (1991). Tumor suppressor genes. Science 254, 1138-1146.
49. Wilson, J. F. (2005). Liver cancer on the rise. Ann Intern Med 142, 1029-
1032.
50. Wooster, R., and Weber, B. L. (2003). Breast and ovarian cancer. N Engl J
Med 348,
2339-2347. Yanaihara, N., Caplen, N., Bowman, E., Kumamoto, K., Okamoto, A.,
Yokota,
J., Tanaka, T.,
51. Calin, G. A., Liu, C. G., Croce, C. M., and Harris, C. C. (2006). microRNA
Signature
in
52. Lung Cancer Diagnosis and Prognosis. Cancer Cell 9, 189-198.
53. Zhang, L., and al, e. (2006). MicroRNAs exhibit high frequency genomic
alterations
in human cancer. Proc Natl Acad Sc U S A 103, 9136-9141.
References for Example II
54. Bejerano, G., Pheasant, M., Makunin, I., Stephen, S., Kent, W. J.,
Mattick, J. S., and
Haussler, D. (2004). Ultraconserved elements in the human genome. Science 304,
1321-
1325.
55. Benjamini Y and Hochberg Y. Controlling the False Discovery Rate: a
Practical and
Powerful Approach to Multiple Testing. Journal of the Royal Statistical
Society. B, 57, 289-
300.
56. Calin, G. A., Sevignani, C., Dumitru, C. D., Hyslop, T., Noch, E.,
Yendamuri, S.,
Shimizu, M., Rattan, S., Bullrich, F., Negrini, M., and Croce, C. M. (2004).
Human
microRNA genes are frequently located at fragile sites and genomic regions
involved in
cancers. Proc Natl Acad Sci U S A 101, 2999-3004.
91

Dessin représentatif

Désolé, le dessin représentatif concernant le document de brevet no 2695514 est introuvable.

États administratifs

2024-08-01 : Dans le cadre de la transition vers les Brevets de nouvelle génération (BNG), la base de données sur les brevets canadiens (BDBC) contient désormais un Historique d'événement plus détaillé, qui reproduit le Journal des événements de notre nouvelle solution interne.

Veuillez noter que les événements débutant par « Inactive : » se réfèrent à des événements qui ne sont plus utilisés dans notre nouvelle solution interne.

Pour une meilleure compréhension de l'état de la demande ou brevet qui figure sur cette page, la rubrique Mise en garde , et les descriptions de Brevet , Historique d'événement , Taxes périodiques et Historique des paiements devraient être consultées.

Historique d'événement

Description Date
Inactive : CIB expirée 2019-01-01
Demande non rétablie avant l'échéance 2018-08-06
Le délai pour l'annulation est expiré 2018-08-06
Requête pour le changement d'adresse ou de mode de correspondance reçue 2018-01-12
Inactive : CIB expirée 2018-01-01
Inactive : Abandon. - Aucune rép dem par.30(2) Règles 2017-12-12
Réputée abandonnée - omission de répondre à un avis sur les taxes pour le maintien en état 2017-08-04
Inactive : Dem. de l'examinateur par.30(2) Règles 2017-06-12
Inactive : Rapport - Aucun CQ 2017-05-24
Modification reçue - modification volontaire 2016-08-19
Inactive : Dem. de l'examinateur par.30(2) Règles 2016-02-19
Inactive : Rapport - CQ échoué - Mineur 2016-02-17
Modification reçue - modification volontaire 2015-06-12
Inactive : Dem. de l'examinateur par.30(2) Règles 2014-12-12
Inactive : Rapport - Aucun CQ 2014-11-28
Lettre envoyée 2013-06-18
Requête d'examen reçue 2013-06-10
Exigences pour une requête d'examen - jugée conforme 2013-06-10
Toutes les exigences pour l'examen - jugée conforme 2013-06-10
Modification reçue - modification volontaire 2013-06-10
Inactive : CIB désactivée 2011-07-29
Inactive : CIB du SCB 2011-01-10
Inactive : CIB expirée 2011-01-01
Inactive : CIB en 1re position 2010-04-29
Inactive : CIB attribuée 2010-04-29
Inactive : CIB attribuée 2010-04-29
Inactive : CIB attribuée 2010-04-29
Inactive : CIB attribuée 2010-04-29
Inactive : CIB enlevée 2010-04-29
Inactive : CIB attribuée 2010-04-29
Inactive : CIB attribuée 2010-04-29
Inactive : CIB attribuée 2010-04-29
Inactive : CIB attribuée 2010-04-29
Inactive : Page couverture publiée 2010-04-26
Inactive : Notice - Entrée phase nat. - Pas de RE 2010-04-09
Inactive : CIB en 1re position 2010-04-06
Inactive : CIB attribuée 2010-04-06
Inactive : CIB attribuée 2010-04-06
Inactive : CIB attribuée 2010-04-06
Demande reçue - PCT 2010-04-06
Exigences pour l'entrée dans la phase nationale - jugée conforme 2010-02-03
Demande publiée (accessible au public) 2009-02-12

Historique d'abandonnement

Date d'abandonnement Raison Date de rétablissement
2017-08-04

Taxes périodiques

Le dernier paiement a été reçu le 2016-07-20

Avis : Si le paiement en totalité n'a pas été reçu au plus tard à la date indiquée, une taxe supplémentaire peut être imposée, soit une des taxes suivantes :

  • taxe de rétablissement ;
  • taxe pour paiement en souffrance ; ou
  • taxe additionnelle pour le renversement d'une péremption réputée.

Les taxes sur les brevets sont ajustées au 1er janvier de chaque année. Les montants ci-dessus sont les montants actuels s'ils sont reçus au plus tard le 31 décembre de l'année en cours.
Veuillez vous référer à la page web des taxes sur les brevets de l'OPIC pour voir tous les montants actuels des taxes.

Historique des taxes

Type de taxes Anniversaire Échéance Date payée
Taxe nationale de base - générale 2010-02-03
TM (demande, 2e anniv.) - générale 02 2010-08-04 2010-07-20
TM (demande, 3e anniv.) - générale 03 2011-08-04 2011-07-19
TM (demande, 4e anniv.) - générale 04 2012-08-06 2012-08-03
Requête d'examen - générale 2013-06-10
TM (demande, 5e anniv.) - générale 05 2013-08-05 2013-07-18
TM (demande, 6e anniv.) - générale 06 2014-08-04 2014-07-22
TM (demande, 7e anniv.) - générale 07 2015-08-04 2015-07-21
TM (demande, 8e anniv.) - générale 08 2016-08-04 2016-07-20
Titulaires au dossier

Les titulaires actuels et antérieures au dossier sont affichés en ordre alphabétique.

Titulaires actuels au dossier
THE OHIO STATE UNIVERSITY RESEARCH FOUNDATION
Titulaires antérieures au dossier
CARLO M. CROCE
Les propriétaires antérieurs qui ne figurent pas dans la liste des « Propriétaires au dossier » apparaîtront dans d'autres documents au dossier.
Documents

Pour visionner les fichiers sélectionnés, entrer le code reCAPTCHA :



Pour visualiser une image, cliquer sur un lien dans la colonne description du document (Temporairement non-disponible). Pour télécharger l'image (les images), cliquer l'une ou plusieurs cases à cocher dans la première colonne et ensuite cliquer sur le bouton "Télécharger sélection en format PDF (archive Zip)" ou le bouton "Télécharger sélection (en un fichier PDF fusionné)".

Liste des documents de brevet publiés et non publiés sur la BDBC .

Si vous avez des difficultés à accéder au contenu, veuillez communiquer avec le Centre de services à la clientèle au 1-866-997-1936, ou envoyer un courriel au Centre de service à la clientèle de l'OPIC.


Description du
Document 
Date
(yyyy-mm-dd) 
Nombre de pages   Taille de l'image (Ko) 
Dessins 2010-02-02 70 5 419
Description 2010-02-02 91 5 189
Revendications 2010-02-02 13 497
Abrégé 2010-02-02 1 50
Page couverture 2010-04-25 1 28
Revendications 2015-06-11 2 70
Description 2015-06-11 91 5 034
Description 2016-08-18 91 5 028
Revendications 2016-08-18 2 91
Rappel de taxe de maintien due 2010-04-06 1 115
Avis d'entree dans la phase nationale 2010-04-08 1 197
Rappel - requête d'examen 2013-04-07 1 119
Accusé de réception de la requête d'examen 2013-06-17 1 177
Courtoisie - Lettre d'abandon (R30(2)) 2018-01-22 1 166
Courtoisie - Lettre d'abandon (taxe de maintien en état) 2017-09-14 1 171
PCT 2010-02-02 2 117
Correspondance 2010-03-16 2 51
Taxes 2010-07-19 1 36
Modification / réponse à un rapport 2015-06-11 35 1 698
Demande de l'examinateur 2016-02-18 4 286
Modification / réponse à un rapport 2016-08-18 9 335
Demande de l'examinateur 2017-06-11 5 210